CA2441792C - Carboline derivatives as inhibitors of phosphodiesterase 5 (pdes) for the treatment of cardiovascular diseases and erectile dysfunction - Google Patents

Carboline derivatives as inhibitors of phosphodiesterase 5 (pdes) for the treatment of cardiovascular diseases and erectile dysfunction Download PDF

Info

Publication number
CA2441792C
CA2441792C CA2441792A CA2441792A CA2441792C CA 2441792 C CA2441792 C CA 2441792C CA 2441792 A CA2441792 A CA 2441792A CA 2441792 A CA2441792 A CA 2441792A CA 2441792 C CA2441792 C CA 2441792C
Authority
CA
Canada
Prior art keywords
group
compound
4alkylenec
6alkyl
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2441792A
Other languages
French (fr)
Other versions
CA2441792A1 (en
Inventor
Jason Scott Sawyer
Mark W. Orme
James D. Copp
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lilly Icos LLC
Original Assignee
Lilly Icos LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lilly Icos LLC filed Critical Lilly Icos LLC
Publication of CA2441792A1 publication Critical patent/CA2441792A1/en
Application granted granted Critical
Publication of CA2441792C publication Critical patent/CA2441792C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Abstract

Compounds of general structural formula (I) and use of the compounds and salts and solvates thereof, as therapeutic agents as inhibitors of phosphodiesterase 5 cardiovascular disorders.

Description

CHEMICAL COMPOUNDS
FIELD AND BACKGROUND OF THE INVENTION
This invention relates to a series of com-pounds, to methods of preparing the compounds, to pharmaceutical compositions containing the com-pounds, and to their use as therapeutic agents. In particular, the invention relates to compounds that are potent and selective inhibitors of cyclic guano-sine 3',5'-monophosphate specific phosphodiesterase (cGMP-specific PDE), in particular PDE5, and have utility in a variety of therapeutic areas wherein such inhibition is considered beneficial, including the treatment of cardiovascular disorders and erec-tile dysfunction.
SUMMARY OF THE INVENTION
The present invention provides compounds of formula (I) ~R~) q X) n-Y
wherein R°, independently, is selected from the group consisting of halo, C1_6alkyl, aryl, heter-oaryl , C3_ecycloalkyl , C3_8heterocycloalkyl , C3_8cyclo-alkylQ, C (=O) Ra, OC (=O) Ra, C (=O) ORa, C1_4alkyleneNRaRb, C1_4alkyleneHet, C1_4alkyleneC (=O) ORa, C (=O) NRaSO2R°, C (=O) C1_4alkyleneHet, C (=O) NRaRb, C (=O) NRaR', C (=O) -NRaCl_4alkyleneORb, C (=O) NRaCl_4alkyleneHet, ORa, OC1_4-alkyleneC (=0) ORa, OC1_4alkyleneNRaRb, OC1_4alkyleneHet, OC1_4alkyleneORa, OC1_4alkyleneNRaC (=O) ORb, NRaRb, NRaCl_4alkyleneNRaRb, NRaC (=O) Rb, NRaC (=O) NRaRb, N (SOzCl_4alkyl) 2, NRa (S02C1_4alkyl) , nitro, trifluoro-methyl, trifluoromethoxy, cyano, SOZNRaRb, SOZRa, SORa, SRa, and OSOzCF3;
R1 is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, an optionally substituted C3_8cycloalkyl ring, an optionally substituted C3_8heterocycloalkyl ring, an optionally substituted bicyclic ring a , wherein the fused ring A is a 5- or 6-membered ring, saturated or partially or fully unsaturated, and comprises carbon atoms and optionally one to three heteroatoms selected from oxygen, sulfur, and nitrogen, hydrogen, C1_6alkyl, arylCl_3alkyl, C1_3-alkylenearyl , haloCl_6alkyl , C1_QalkyleneC (=O) ORa, C1_4alkyleneC (=O) NRaRb, C3_8cycloalkyl, C3_ecycloalkenyl, C3_eheterocycloalkenyl, C1_4alkyleneHet, C1_4alkylene-QRa, C2_6alkenyleneQRa, C1_4alkyleneQC1_4alkyleneQRa, D B
/ I ~~Rc C E
D E
/ I ~~Rc C B
D
/ I (R0) q C
and a spiro substituent having a structure O O
SRO) q R2 is selected from the group consisting of hydrogen, C1_salkyl, C3_ecycloalkyl, C3_sheterocyclo-alkyl , Cz_6alkenyl , C1_3alkylenearyl , arylCl_3alkyl , C (=O) Ra, aryl, heteroaryl, C (=O) Ra, C (=O) NRaRb, C (=O) -NRaR°, C (=S) NRaRb, C (=S) NRaR~, S02Ra, SOZNRaRb, S (=O) Ra, S (=O) NRaRb, C (=O) NRaCi_4alkyleneORa, C (=O) NRaCl_4alkyl-eneHet, C (=O) C1_4alkylenearyl, C (=O) C,_4alkylene-heteroaryl, C1_4alkylenearyl substituted with one or more of SOzNRaRb, NRaRb, C (=O) ORa, NRaSOzCF3, CN, NO2, C (=O) Ra, ORa, C,_QalkyleneNRaRb, and OC1_4alkyleneNRaRb, C1_4alkyleneheteroaryl, C1_4alkyleneHet, C1_4alkyleneC-(=O) C1_4alkylenearyl, C1_4alkyleneC (=O) C1_4alkylene-heteroaryl, C1_4alkyleneC (=O) Het, C1_4alkyleneC (=O) -NRaRb, C1_4alkyleneORa, C1_4alkyleneNRaC (=O) Ra, C1_4alkyl-eneOCl_4alkyleneORa, C1_4alkyleneNRaRb, C1_4alkyleneC-(=O) ORa, and C1_4alkyleneOC1_4alkyleneC (=O) ORa;
R3 is selected from the group consisting of C (=O) Rb, C (=O) ORb, C (=O) NRaRb, C (=O) NRaR~, C (=S) NRaRb, C (=S) NRaR°, C (=O) Het, C (=O) NRaCl_4alkyleneORa, C (=O) NRaCl_4alkyleneHet, C (=O) C1_4alkylenearyl, C (=O) C1_Qalkyleneheteroaryl, C (=O) NRaCl_4alkylenearyl, C (=0) NRaCl_4alkyleneC3_ecycloalkyl, C (=O) NRbS02R°, C (=0) NRaCl_4alkyleneOC1_6alkyl, C (=O) NRaCl_4alkylene-heteroaryl, NRaR°, NRaC (=O) Rb, NRaC (=O) NRaR°, NRa (SOZC1_4alkyl) , N (SOzCl_4alkyl ) 2, ORa, NRaC (=O) C1_4alkyleneN (Rb) 2, NRaC (=O) C1_4alkyleneC (=O) ORa, NRa (C=O) C1_3alkylenearyl, NRaC (=0) C1_3alkylene-C3_$heterocycloalkyl, NRaC (=0) C1_3alkyleneHet, and C (=0) NRaSO2Rb;
R' is selected from the group consisting of hydrogen, C1_6alkyl , aryl , heteroaryl , arylCl_3alkyl , C1_3alkylenearyl, C1_3alkyleneHet, C3_ecycloalkyl, and C3_eheterocycloalkyl ;
X is selected from the group consisting of C (=O) , (CHz) tC (=O) , C (=O) C=C, C (=O) C (Ra) =C (Rd) , C (=S) , SO, SO2, SOzC (Ra) =CRa, CRaRb, CR''=CRa, C (=O) NRa, arid C (=N-ORa) ;
Y is selected from the group consisting of Ra i Rd i ( CHZ ) nC ( =O ) R~ i N ( Rb ) ( CH2 ) aR' i O ( CH2 ) nR~ i N (Rb) C (=O) R~, C (=O) N (Ra) (R~) , N (Ra) C (=O) R~, CRa and N ( Ra ) SOzR° ;
Ra is selected from the group consisting of hydrogen, C1_6alkyl, C3_6cycloalkyl, aryl, arylCl_3alkyl, C1_3alkylenearyl, heteroaryl, heteroarylCl_3alkyl, and C1_3alkyleneheteroaryl;
Rb is selected from the group consisting of hydrogen, C1_6alkyl, C3_$cycloalkyl, C1_3alkyleneN(Ra)z, aryl , arylCl_3alkyl , C1_3alkylenearyl , and heteroaryl ;
R~ is selected from the group consisting of 2 0 hydrogen, C1_6alkyl , aryl , heteroaryl , arylCl_3alkyl , heteroarylCl_3alkyl, C1_3alkyleneN (Ra) z, C1_6alkylene-aryl, C1_6alkyleneHet, haloCl_6alkyl, C3_8cycloalkyl, C3_eheterocycloalkyl, Het, C1_3alkyleneheteroaryl, C1_6alkyleneC (=O) ORa, and C1_3alkyleneC3_eheterocyclo-alkyl;
or Ra and R° are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;
Rd is a 5- or 6-membered ring or a bicyclic fused ring system, saturated or partially or fully unsaturated, comprising carbon atoms and optionally one to three heteroatoms selected from oxygen, sulfur, and nitrogen, and optionally substituted with one or more Re or Rf ;
Re is selected from the group consisting of - nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, Het, C1_6alkyl, C1_6alkyleneORa, C (=O) R'', OC ( =O ) Ra , C ( =O ) ORa , C1_4alkyleneHet , C1_4alkyleneC (=O) ORa, OC1_4alkyleneC (=O) ORa, C1_4alkyleneOC1_4alkyleneC (=O) ORa, C ( =O ) NRaSOzRf , C (=O) C1_4alkyleneHet, C1_4alkyleneNRaRb, CZ_6alkenyleneNRaRb, C ( =O ) NRaRb , C (=O) NRaRg, C (=O) NRaCl_4alkyleneORb, C (=O) NRaCl_4alkyleneHet, OR' , OC2_4alkyleneNRaRb, OC1_4alkyleneCH (ORa) CHZ-NRaRb, 3 0 OC1_4alkyleneHet , OCZ_4alkyleneORa, OC2_4alkyleneNRaC (=O) ORb, NRaRb , NRaCl_QalkyleneNRaRb, NRaC (=0) Rb, NRaC ( =O ) NRaRb , N (SOZC1_4alkyl ) 2, NRa ( S02C1_4alkyl ) , SO2NRaRb , and OSOztrifluoromethyl;
Rf is selected from the group consisting of hydrogen, halogen, ORa, C1_6alkyl, nitro, and NRaRb;
or Re and Rf are taken together to form a 3- or 4-membered alkylene or alkenylene chain component of a 5- or 6-membered ring, optionally containing at least one heteroatom;
Rg is phenyl or C4_6cycloalkyl, optionally substituted with one or more halogen, C(=O)ORa, or ORa ;
Q is O, S, or NR'';
B is O, S, or NR'';
C is O, S, Or NRa;
D is CRa Or N;
E is CRa, C (Ra) z, or NRh; and R'' is null or is selected from the group consisting of hydrogen, C1_6alkyl, aryl, heteroaryl, arylCl_3alkyl, heteroarylCl_3alkyl, C1_3alkylenearyl, and C1_3alkyleneheteroaryl;
Het represents a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1_4alkyl or C (=0) ORa;
n is 0 or 1;
q is 0, l, 2, 3, or 4;

- g _ t is 1, 2, 3, or 4;
and pharmaceutically acceptable salts and solvates (e. g., hydrates) thereof.
The present invention also provides com-pounds of structural formula (Ia), i.e., compounds of structural formula (I) wherein (X)n-Y is hydrogen, i.e., a compound having a formula *~
~R~)q N * NCH
I

wherein R°, independently, is selected from the group consisting of halo, C1_6alkyl, aryl, heter-oaryl , C3_8cycloalkyl , C3_aheterocycloalkyl , C3_$cyclo-alkylQ, C (=0) Ra, OC (=O) Ra, C (=O) ORa, C1_4alkyleneNRaRb, C1_4alkyleneHet, C1_4alkyleneC (=O) ORa, C (=O) NRaSOZR~, C (=0) C1_4alkyleneHet, C (=O) NRaRb, C (=O) NRaR°, C (=O) -NRaCl_4alkyleneORb, C(=O)NRaCl_4alkyleneHet, ORa, OC1_4-alkyleneC (=0) ORa, OC1_4alkyleneNRaRb, OC1_4alkyleneHet, OC1_4alkyleneORa, OC1_4alkyleneNRaC (=O) ORb, NRaRb, NRaCl_4alkyleneNRaRb, NRaC (=O) Rb, NRaC (=O) NRaRb, N(S02C1_4alkyl)2, NRa(SOzCI_4alkyl) , nitro, trifluoro-methyl, trifluoromethoxy, cyano, SOZNRaRb, SOZRa, SORa, SRa, and OSOZCF3;
R1 is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, an optionally substituted C3_ecycloalkyl ring, an optionally substituted C3_eheterocycloalkyl ring, an optionally substituted bicyclic ring / I A
wherein the fused ring A is a 5- or 6-membered ring, saturated or partially or fully unsaturated, and comprises carbon atoms and optionally one to. three heteroatoms selected from oxygen, sulfur, and nitrogen, hydrogen, C1_6alkyl, arylCl_3alkyl, C1_3 alkylenearyl, haloCl_6alkyl, C1_4alkyleneC (=O) ORa, C1_4alkyleneC(=O)NRaRb, C3_8cycloalkyl, C3_ecycloalkenyl, C3_8heterocycloalkenyl, C1_4alkyleneHet, C1_4alkylene-QRa, Cz_6alkenyleneQRa, C1_4alkyleneQC1_4alkyleneQRa, D B
/ I ~~Rc C E
D E
/ I ~~Rc C B

D
(R0) q C
and a spiro substituent having a structure O O

Rz is selected from the group consisting of hydrogen, C1_6alkyl, C3_ecycloalkyl, C3_8heterocyclo-alkyl, Cz_6alkenyl, C1_3alkylenearyl, arylCl_3alkyl, C (=O) Ra, aryl, heteroaryl, C (=O) Ra, C (=O) NRaRb, C (=O) -NRaR~, C (=S) NRaRb, C (=S) NRaR~, SOZRa, SOZNRaRb, S (=O) Ra, S (=O) NRaRb, C (=O) NRaCl_4alkyleneORa, C (=O) NRaCl_4alkyl-eneHet, C (=0) C1_Qalkylenearyl, C (=O) C1_4alkylene-heteroaryl, C1_4alkylenearyl substituted with one or more of SOzNRaRb, NRaRb, C (=O) ORa, NRaSO2CF3, CN, NO2, C (=O) Ra, ORa, C1_4alkyleneNRaRb, and OC1_4alkyleneNRaRb, C1_4alkyleneheteroaryl, C1_4alkyleneHet, C1_4alkyleneC-(=O) C1_qalkylenearyl, C1_QalkyleneC (=O) C1_4alkylene-heteroaryl, C1_4alkyleneC (=O) Het, C1_4alkyleneC (=O) -NRaRb, C1_4alkyleneORa, C1_4alkyleneNRaC (=O) Ra, C1_4alkyl-eneOCl_4alkyleneORa, C1_4alkyleneNRaRb, C1_4alkyleneC-(=O) ORa, and C1_4alkylene0C1_4alkyleneC (=O) ORa;

R3 is selected from the group consisting of C (=O) Rb, C (=O) ORb, C (=O) NRaRb, C (=O) NRaR°, C (=S) NRaRb, C (=S) NRaR~, C (=O) Het, C (=O) NRaCl_4alkyleneORa, C (=O) -NRaCI_4alkyleneHet, C (=O) C1_4alkylenearyl, C (=O) C1_9-alkyleneheteroaryl, C (=O) NRaCl_4alkylenearyl, C (=O) -NRaCl_4alkyleneC3_ecycloalkyl, C (=O) NRbSO2R~, C (=O) -NRaCI_4alkyleneOC1_6alkyl, C (=O) NRaCI_4alkylenehetero-aryl, NRaR°, NRaC (=O) Rb, NRaC (=O) NRaR°, NRa (SOzCl_4-alkyl) , N (SOzCI_4alkyl) z, ORa, NRaC (=O) C1_4alkylene-N (Rb) z, NRaC (=O) C1_4alkyleneC (=0) ORa, NRa (C=O) C1_3-alkylenearyl, NRaC (=O) C1_3alkyleneC3_8heterocycloalkyl, NRaC (=O) C1_3alkyleneHet, and C (=O) NRaSOZRb;
R4 is selected from the group consisting of hydrogen, C1_6alkyl , aryl , heteroaryl , arylCl_3alkyl , C1_3alkylenearyl, C1_3alkyleneHet, C3_$cycloalkyl, and C3_eheterocycloalkyl;
Ra is selected from the group consisting of hydrogen, C1_6alkyl , C3_6cycloalkyl , aryl , arylCl_3alkyl, C1_3alkylenearyl, heteroaryl, heteroarylCl_3alkyl, and C1_3alkyleneheteroaryl;
Rb is selected from the group consisting of hydrogen, C1_6alkyl, C3_$cycloalkyl, C1_3alkyleneN (Rd) z, aryl , arylCl_3alkyl , C1_3alkylenearyl , and heteroaryl ;
R° is selected from the group consisting of 2 5 hydrogen, C1_6alkyl , aryl , heteroaryl , arylCi_3alkyl , heteroarylCl_3alkyl, C1_3alkyleneN (Ra) z, C1_6alkylene-aryl, C1_6alkyleneHet, haloCl_6alkyl, C3_8cycloalkyl, C3_eheterocycloalkyl, Het, C1_3alkyleneheteroaryl, C1_6alkyleneC (=O) ORa, and C1_3alkyleneC3_eheterocyclo-alkyl;
or Ra and R~ are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;

Q is O, S, or NR'';
B is O, S, or NRh;
C iS O, S, Or NRa;
D is CRa or N;
E is CRa, C (Ra) 2, or NRh; and R'' is null or is selected from the group consisting of hydrogen, C1_6alkyl, aryl, heteroaryl, arylCl_3alkyl , heteroarylCl_3alkyl ; C1_3alkylenearyl , and C1_3alkyleneheteroaryl ;
Het represents a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1_4alkyl or C (=O) ORa;
n is 0 or 1;
q is 0, 1, 2, 3, or 4;
t is l, 2, 3, or 4;
and pharmaceutically acceptable salts and solvates thereof.
As used herein, the term "alkyl" includes straight chained and branched hydrocarbon groups containing the indicated number of carbon atoms, typically methyl, ethyl, propyl, and butyl groups.
The hydrocarbon group can contain up to 16 carbon atoms. The term "alkyl" also includes "bridged alkyl," i.e., a C4-C16 bicyclic or polycyclic hydro-carbon group, for example, norbornyl, adamantyl, bicyclo[2.2.2]octyl, bicyclo[2.2.1]heptyl, bicyclo-[3.2.1]octyl, or decahydronaphthyl. The term "cycloalkyl" is defined as a cyclic C3-CB hydrocarbon group, e.g., cyclopropyl, cyclobutyl, cyclohexyl, and cyclopentyl.

The term "alkenyl" is defined identically as "alkyl," except for containing a carbon-carbon double bond. "Cycloalkenyl" is defined similarly to cycloalkyl, except a carbon-carbon double bond is present in the ring.
The term "alkylene" refers to an alkyl group having a substituent. For example, the term "C1_3alkylenearyl" refers to an alkyl group contain-ing one to three carbon atoms, and substituted with an aryl group. The term "alkenylene" as used herein is similarly defined, and contains the indicated number of carbon atoms and a carbon-carbon double bond, and includes straight chained and branched alkenylene groups, like ethyenylene.
The term "halo" or "halogen" is defined herein to include fluorine, bromine, chlorine, and iodine.
The term "haloalkyl" is defined herein as an alkyl group substituted with one or more halo substituents, either fluoro, chloro, bromo, iodo, or combinations thereof. Similarly, "halocycloalkyl"
is defined as a cycloalkyl group having one or more halo substituents.
The term "aryl," alone or in combination, is defined herein as a monocyclic or polycyclic aromatic group, preferably a monocyclic or bicyclic aromatic group, e.9., phenyl or naphthyl. Unless otherwise indicated, an "aryl" group can be unsub-stituted or substituted, for example, with one or more, and in particular one to three, halo, alkyl, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkyl, nitro, amino, alkylamino, acylamino, alkylthio, alkylsul-finyl, and alkylsulfonyl. Exemplary aryl groups include phenyl, naphthyl, tetrahydronaphthyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2-methylphenyl, 4-methoxyphenyl, 3-trifluoromethyl-phenyl, 4-nitrophenyl, and the like. The terms "arylCl_3alkyl" and "heteroarylCl_3alkyl" are defined as an aryl or heteroaryl group having a C1_3alkyl substituent.
The term "heteroaryl" is defined herein as a monocyclic or bicyclic ring system containing one or two aromatic rings and containing at least one nitrogen, oxygen, or sulfur atom in an aromatic ring, and which can be unsubstituted or substituted, for example, with one or more, and in particular one to three, substituents, like halo, alkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, haloalkyl, nitro, amino, alkylamino, acylamino, alkylthio, alkyl-sulfinyl, and alkylsulfonyl. Examples of heteroaryl groups include thienyl, furyl, pyridyl, oxazolyl, quinolyl, isoquinolyl, indolyl, triazolyl, isothia-zolyl, isoxazolyl, imidizolyl, benzothiazolyl, pyrazinyl, pyrimidinyl, thiazolyl, and thiadiazolyl.
The term "heterocycloalkyl" is defined as monocyclic, bicyclic, and tricyclic groups contain-ing one or more, e.g., one to three, heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur. A "heterocycloalkyl"
group also can contain an oxo group (=0) attached to the ring. Nonlimiting examples of heterocycloalkyl groups include 1,3-dioxolane, 2-pyrazoline, pyrazolidine, pyrrolidine, piperazine, a pyrroline, 2H-pyran, 4H-pyran, morpholine, thiopholine, piperidine, 1,4-dithiane, and 1,4-dioxane.
The term "hydroxy" is defined as -OH.

The term "alkoxy" is defined as -OR, wherein R is alkyl.
The term "alkoxyalkyl" is defined as an alkyl group wherein a hydrogen has been replaced by an alkoxy group. The term "(alkylthio)alkyl" is defined similarly as alkoxyalkyl, except a sulfur atom, rather than an oxygen atom, is present.
The term "hydroxyalkyl" is defined as a hydroxy group appended to an alkyl group.
The term "amino" is defined as -NH2, and the term "alkylamino" is defined as -NR2, wherein at least one R is alkyl and the second R is alkyl or hydrogen.
The term "acylamino" is defined as RC(=O)N, wherein R is alkyl or aryl.
The term "alkylthio" is defined as -SR, wherein R is alkyl.
The term "alkylsulfinyl" is defined as R-SO2, wherein R is alkyl.
The term "alkylsulfonyl" is defined as R-503, wherein R is alkyl.
The term "nitro" is defined as -NO2.
The term "trifluoromethyl" is defined as -CF3.
The term "trifluoromethoxy" is defined as -OCF3 .
The term "spiro" as used herein refers to a group having two carbon atoms directly bonded to the carbon atom to which R1 is attached.
The term "cyano" is defined as -CN.
In a preferred embodiment, q is 0. In other preferred embodiments, R° is selected from the group consisting of aryl, Het, ORa, C (=O) ORa, C1_4-alkyleneNRaRb, OC (=O) Ra, C (=O) Ra, NRaRb, C3_acycloalkyl, C3_ecycloalkylQ, C (=O) NRaRb, and C (=O) NRaR'.
In a preferred group of compounds of formula (I) , R1 is represented by A
wherein the bicyclic ring can represent, for example, naphthalene or indene, or a hetero-cycle, such as benzoxazole, benzothiazole, benzi-soxazole, benzimidazole, quinoline, indole, benzo-thiophene, or benzofuran, or G~
(CH2)p G
wherein p is an integer 1 or 2, and G, independent-ly, is C (Ra) z, O, S, or NRa. The bicyclic ring com-prising the R1 substituent typically is attached to the rest of the molecule by a phenyl ring carbon atom.
In another preferred group of compounds of formula (I), R1 is represented by an optionally sub stituted bicyclic ring / G\
(CH2 ) P
~G
, wherein p is 1 or 2, and G, independently, are C(Ra)z or O.
Especially preferred R1 substituents include \ N

10 \

O
\
O
O
and O
-Within this particular group of compounds, nonlimit-ing examples of substituents for the bicyclic ring include halogen (e. g., chlorine), C1_3alkyl (e. g., methyl, ethyl, or i-propyl), ORa (e. g., methoxy, ethoxy, or hydroxy), COZRa, halomethyl or halomethoxy (e. g., trifluoromethyl or trifluoromethoxy), cyano, ni t ro , and NRaRb .
In other preferred embodiments, R1 is optionally substituted and selected from the group consisting of C1_4alkyleneQRa, C1_4alkyleneQC1_4alkyl-eneQRa, C3_ecycloalkyl, C3_scycloalkenyl, C1_6alkyl, D B
~ ,~---R
~C E
D E
~ ,~-R
B
D \
(R0)q C

(R~) q In a more preferred group of compounds of formula (I), R1 is represented by D B
/ ~ ~~Rc ~C E
D E
/ ~ /~Rc ~C B
D
(R0)q C
C3_$cycloalkyl, C3_8cycloalkenyl, C1_6alkyl, C1_4alkyl-eneQRa, and C1_4alkyleneQC1_4alkyleneQRa. A preferred Q is oxygen.
Some preferred R1 substituents are S
O
, N
I
Ra , S O
N N
I I
Ra Ra , -CH20Ra, -CHzOCH~ORa, and Within this particular group of compounds, preferred Ra substituents include hydrogen, C1_6alkyl, and benzyl.
In a preferred embodiment, RZ is selected from the group consisting of hydrogen, aryl, hetero-aryl, ORa, NRaRb, NRaR°, C,_4alkyleneHet, C1_4alkylene-heteroaryl, C1_4alkylenearyl, C1_QalkyleneC (=O) C1_4-alkylenearyl, C1_4alkyleneC (=O) ORa, C1_4alkyleneC (=O) -NRaRb, C1_4alkyleneC (=O) NRaR~, C1_4alkyleneC (=O) Het, C1_4alkyleneNRaRb, C1_4alkyleneNRaR°, C1_4alkyleneNRaC-(=O)Ra, and C1_4alkyleneOC1_4alkyleneORa.
In more preferred embodiments, RZ is selected from the group consisting of hydrogen; C1_4-alkyleneheteroaryl, wherein the heteroaryl group is selected from the group consisting of benzimidazole, a triazole, and imidazole; C1_4alkyleneHet, wherein Het is selected from the group consisting of piper-azine, morpholine, pyrrolidine, pyrrolidone, tetra-hydrofuran, piperidine, and C1_4alkyleneC6H5, optionally substituted with one to three groups selected from the group consisting of C (=O) ORa, NRaRb, NR~SOZCF3, SOzNRaRb, CN, ORa, C (=O) Ra, C1_4alkyleneNRaRb, nitro, OC1_4alkylenearyl, and OC1_4alkyleneNRaRb; C1_4alkyleneC (=O) benzyl; C1_4alkyl-eneC (=O) ORa; C1_4alkyleneC (=O) NRaRb; C1_4alkyleneC (=O) -NRaR~; C1_4alkyleneHet; NRaRb; OH; OC1_4alkyl; C6H5;
C1_4alkyleneNRaRb; C1_4alkyleneORa; C1_4alkyleneNHC
(=O) Ra; and C1_4alkyleneOC1_4alkyleneORa. In most preferred embodiments, RZ is hydrogen.
In preferred embodiments, R3 is selected from the group consisting of C (=O) ORa, C (=O) Ra, C (=O) NRaCl_4alkyleneOC1_6alkyl, C (=O) NRaCl_4alkylene-C3_acycloalkyl, C (=O) Het, C (=O)NRaCl_4alkylene A
C (=0) NRaCl_4alkyleneheteroaryl, C (=O) NRaCi_4alkylene-aryl, C (=O) NRaCl_4alkyleneHet, C (=O) NRaR~, and C (=S) NRaR°;
In preferred embodiments, R4 is selected from the group consisting of hydrogen, C1_6alkyl, aryl, and heteroaryl.
In preferred embodiments, n is 0, or X is selected from the group consisting of C(=O), C (=0) CRa=CRa, (CHZ) tC=O, C (=S) , arid C (=N-ORa) .
In preferred embodiments, Y is selected from the group consisting of Ra, Rd, N (Rb) (CHz) "Rc, O (CHZ) nR', N (Rb) C (=O) R~, and N (Rd) C (=O) R~.

In especially preferred embodiments, q is 0 or R° is selected from the group consisting of halo, methyl, trifluoromethyl, and trifluoromethyl;
R1 is selected from the group consisting of O
O~ , /
O

, W
N

Cl and RZ is selected from the group consisting of hydrogen, C1_6alkyl, C (=O) NRaR°, and C14alkyleneHet; R3 is selected from the group consisting of C(=O)OCZHS, C ( =O ) OCH3 , C ( =O ) NHCHzC6H5 , C ( =O ) NH ( CHZ ) 2C6Hs , C (=O) NHC6H5, C (=O) NH2, C (=O) N (CH3) 2, C (=S) N (CH3) 2, C ( =O ) NH ( CHZ ) ZCH3 , C ( =O ) N ( CHZ ) 3CH3 , C ( =O ) NHCH3 , C ( =0 ) NHCH ( CH3 ) 2 , C ( =O ) NH ( CHZ ) 30CH3 , C ( =O ) NHCH 2 C (=O) NH
C(=O)NH(CH2)3-N O

C (=O) NH (CH2) 3-N
~N
C (=O) NH (CH2) 2 ~ ~ OCH3 C (=0) NH ~ ~ F
C(=O)NHCH2 ~ ~ OCH3 O
C(=O)NHCH2 ~O

C(=O)NH--a C ( =O ) NHCH 2 C ( =O ) NHCH 2 ~ ~ F
C(=O)NH(CH2)2 ~ ~ F
C (=O) -N CH3 C (=0) NH (CH2) 2 j ~ ~N
C ( =O ) NHCH 2 C(=O)NH(CHz)2-N O
C ( =O ) NHCH 2 \
~NHC ( =O ) CH 3 C ( =O ) NH ( CH 2 ) 2 v ~OCH3 C(=O)NH(CH2)3-N
p and C(=O)NHCH2 R' is selected from the group consisting of hydrogen and C16alkyl; X is selected from the group consist-ing of C(=O), C(=S), CHzC(=O), and C(=O)CH=CH, or n is 0; and Y is selected from the group consisting of NH ( CHZ ) 3CH3 , OCH3 , C6H5 , and O

An especially preferred subclass of com-pounds within the general scope of formula (I) is represented by compounds of formula (II) (R~) q (X) n_Y

(II) and pharmaceutically acceptable salts and solvates (e. g., hydrates) thereof.
Compounds of formula (I) can contain one or more asymmetric center, and, therefore, can exist as stereoisomers. The present invention includes both mixtures and separate individual stereoisomers of the compounds of formula (I). Compounds of formula (I) also can exist in tautomeric forms, and the invention includes both mixtures and separate individual tautomers thereof.
Pharmaceutically acceptable salts of the compounds of formula (I) can be acid addition salts formed with pharmaceutically acceptable acids.
Examples of suitable salts include, but are not limited to, the hydrochloride, hydrobromide, sul-fate, bisulfate, phosphate, hydrogen phosphate, ace-tate, benzoate, succinate, fumarate, maleate, lac-tate, citrate, tartrate, gluconate, methanesul-fonate, benzenesulfonate, and p-toluenesulfonate salts. The compounds of formula (I) also can provide pharmaceutically acceptable metal salts, in particular alkali metal salts and alkaline earth metal salts, with bases. Examples include the sodium, potassium, magnesium, and calcium_salts.
Compounds of the present invention are potent and selective inhibitors of cGMP-specific PDES. Thus, compounds of formula (I) are of interest for use in therapy, specifically for the treatment of a variety of conditions where selective inhibition of PDE5 is considered to be beneficial.
Phosphodiesterases (PDEs) catalyze the hydrolysis of cyclic nucleotides, such as cyclic adenosine monophosphate (CAMP) and cyclic guanosine monophosphate (cGMP). The PDEs have been classified into at least seven isoenzyme families and are present in many tissues (J. A. Beavo, Physiol. Rev., 75, p. 725 (1995)).
PDE5 inhibition is a particularly attrac-tive target. A potent and selective inhibitor of PDE5 provides vasodilating, relaxing, and diuretic effects, all of which are beneficial in the treat-ment of various disease states. Research in this area has led to several classes of inhibitors based on the cGMP basic structure (E. Sybertz et al., Expert. Opin. Ther. Pat., 7, p. 631 (1997)).
The biochemical, physiological, and clini-cal effects of PDE5 inhibitors therefore suggest their utility in a variety of disease states in which modulation of smooth muscle, renal, hemostat-ic, inflammatory, and/or endocrine function is de-sirable. The compounds of formula (I), therefore, have utility in the treatment of a number of dis-orders, including stable, unstable, and variant (Prinzmetal) angina, hypertension, pulmonary hyper-tension, congestive heart failure, acute respiratory distress syndrome, acute and chronic renal failure, atherosclerosis, conditions of reduced blood vessel patency (e. g., postpercutaneous transluminal coro-nary or carotid angioplasty, or post-bypass surgery graft stenosis), peripheral vascular disease, vas-cular disorders, such as Raynaud's disease, thrombo-cythemia, inflammatory diseases, stroke, bronchitis, chronic asthma, allergic asthma, allergic rhinitis, glaucoma, osteoporosis, preterm labor, benign pros-tatic hypertrophy, peptic ulcer, male erectile dys-function, female sexual dysfunction, and diseases characterized by disorders of gut motility (e. g., irritable bowel syndrome).
An especially important use is the treat-ment of male erectile dysfunction, which is one form of impotence and is a common medical problem. Impo-tence can be defined as a lack of power, in the male, to copulate, and can involve an inability to achieve penile erection or ejaculation, or both.
The incidence of erectile dysfunction increases with age, with about 500 of men over the age of 40 suf-fering from some degree of erectile dysfunction.
In addition, a further important use is the treatment of female arousal disorder. Female arousal disorders are defined as a recurrent in-ability to attain or maintain an adequate lubrica-tion/swelling response of sexual excitement until completion of sexual activity. The arousal response consists of vasocongestion in the pelvis, vaginal lubrication, and expansion and swelling of external genitalia.
It is envisioned, therefore, that com-pounds of formula (I) are useful in the treatment of male erectile dysfunction and female arousal dis-order. Thus, the present invention concerns the use of compounds of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical compo-sition containing either entity, for the manufacture of a medicament for the curative or prophylactic treatment of erectile dysfunction in a male animal and arousal disorder in a female animal, including humans.
The term "treatment" includes preventing, lowering, stopping, or reversing the progression or severity of the condition or symptoms being treated.
As such, the term "treatment" includes both medical therapeutic and/or prophylactic administration, as appropriate.
It also is understood that "a compound of formula (I)," or a physiologically acceptable salt or solvate thereof, can be administered as the neat compound, or as a pharmaceutical composition con-taining either entity.
Although the compounds of the invention are envisioned primarily for the treatment of sexual dysfunction in humans, such as male erectile dys-function and female arousal disorder, they also can be used for the treatment of other disease states.
A further aspect of the present invention, therefore, is providing a compound of formula (I) for use in the treatment of stable, unstable, and variant (Prinzmetal) angina, hypertension, pulmonary hypertension, chronic obstructive pulmonary disease, congestive heart failure, acute respiratory distress syndrome, acute and chronic renal failure, athero-sclerosis, conditions of reduced blood vessel paten-cy (e. g., post-PTCA or post-bypass graft stenosis), peripheral vascular disease, vascular disorders such as Raynaud's disease, thrombocythemia, inflammatory diseases, prophylaxis of myocardial infarction, prophylaxis of stroke, stroke, bronchitis, chronic asthma, allergic asthma, allergic rhinitis, glau-coma, osteoporosis, preterm labor, benign prostatic hypertrophy, male and female erectile dysfunction, or diseases characterized by disorders of gut motility (e. g., IBS).

According to another aspect of the present invention, there is provided the use of a compound of formula (I) for the manufacture of a medicament for the treatment of the above-noted conditions and disorders.
In a further aspect, the present invention provides a method of treating the above-noted con-ditions and disorders in a human or nonhuman animal body which comprises administering to said body a therapeutically effective amount of a compound of formula (I).
Compounds of the invention can be admin-istered by any suitable route, for example by oral, buccal, inhalation, sublingual, rectal, vaginal, transurethral, nasal, topical, percutaneous, i.e., transdermal, or parenteral (including intravenous, intramuscular, subcutaneous, and intracoronary) administration. Parenteral administration can be accomplished using a needle and syringe, or using a high pressure technique, like POWDERJECTT"".
Oral administration of a compound of the invention is the preferred route. Oral administra-tion is the most convenient and avoids the dis-advantages associated with other routes of admin-istration. For patients suffering from a swallowing disorder or from impairment of drug absorption after oral administration, the drug can be administered parenterally, e.g., sublingually or buccally.
Compounds and pharmaceutical compositions suitable for use in the present invention include those wherein the active ingredient is administered in an effective amount to achieve its intended pur-pose. More specifically, a "therapeutically effec-tive amount" means an amount effective to prevent development of, or to alleviate the existing symp-toms of, the subject being treated. Determination of the effective amounts is well within the cap-s ability of those skilled in the art, especially in light of the detailed disclosure provided herein.
A "therapeutically effective dose" refers to that amount of the compound that results in achieving the desired effect. Toxicity and thera-peutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cul-tures or experimental animals, e.g., for determining the LDso (the dose lethal to 50% of the population) and the EDSO (the dose therapeutically effective in 500 of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LDso and EDso. Compounds which exhibit high therapeutic indices are preferred. The data obtained from such data can be used in formulating a range of dosage for use in humans. The dosage of such compounds preferably lies within a range of circulating con-centrations that include the EDso with little or no toxicity. The dosage can vary within this range depending upon.the dosage form employed, and the route of administration utilized.
The exact formulation, route of admin-istration, and dosage can be chosen by the indi-vidual physician in view of the patient's condition.
Dosage amount and interval can be adjusted individ-ually to provide plasma levels of the active moiety which are sufficient to maintain the therapeutic effects.

The amount of composition administered is dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration, and the judgment of the prescribing physician.
Specifically, for administration to a human in the curative or prophylactic treatment of the conditions and disorders identified above, oral dosages of a compound of formula (I) generally are about 0.5 to about 1000 mg daily for an average adult patient (70 kg). Thus, for a typical adult patient, individual tablets or capsules contain 0.2 to 500 mg of active compound, in a suitable pharma-ceutically acceptable vehicle or carrier, for ad-ministration in single or multiple doses, once or several times per day. Dosages for intravenous, buccal, or sublingual administration typically are 0.1 to 500 mg per single-dose as required. In practice, the physician determines the actual dosing regimen which is most suitable for an individual patient, and the dosage varies with the age, weight, and response of the particular patient. The above dosages are exemplary of the average case, but there can be individual instances in which higher or lower dosages are merited, and such are within the scope of this invention.
For human use, a compound of the formula (I) can be administered alone, but generally is ad-ministered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical prac-tice. Pharmaceutical compositions for use in accordance with the present invention thus can be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries that facilitate proces-sing of compounds of formula (I) into preparations which can be used pharmaceutically.
These pharmaceutical compositions can be manufactured in a conventional manner, e.g., by conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulat-ing, entrapping, or lyophilizing processes. Proper formulation is dependent upon the route of admin-istration chosen. When a therapeutically effective amount of a compound of the present invention is administered orally, the composition typically is in the form of a tablet, capsule, powder, solution, or elixir. When administered in tablet form, the com-position can additionally contain a solid carrier, such as a gelatin or an adjuvant. The tablet, cap-sule, and powder contain about 5% to about 95% com-pound of the present invention, and preferably from about 25% to about 90% compound of the present invention. When administered in liquid form, a liquid carrier such as water, petroleum, or oils of animal or plant origin can be added. The liquid form of the composition can further contain physiological saline solution, dextrose or other saccharide solutions, or glycols. When administered in liquid form, the composition contains about 0.5%
to about 90% by weight of a compound of the present invention, and preferably about 1% to about 50% of a compound of the present invention.
When a therapeutically effective amount of a compound of the present invention is administered by intravenous, cutaneous, or subcutaneous injec-tion, the composition is in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable solu-tions, having due regard to pH, isotonicity, stabil-ity, and the like, is within the skill in the art.
A preferred composition for intravenous, cutaneous, or subcutaneous injection typically contains, in addition to a compound of the present invention, an isotonic vehicle.
For oral administration, the compounds can be formulated readily by combining a compound of formula (I) with pharmaceutically acceptable car-riers well known in the art. Such carriers enable the present compounds to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral inges-tion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by adding a compound of formula (I) with a solid excipient, optionally grinding a resulting mixture, and proces-sing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, for example, fillers and cellulose preparations. If desired, disintegrating agents can be added.
For administration by inhalation, com-pounds of the present invention are conveniently delivered in the form of an aerosol spray presen-tation from pressurized packs or a nebulizer, with the use of a suitable propellant. In the case of a pressurized aerosol, the dosage unit can be deter-mined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin, for use in an inhaler or insufflator can be formu-lated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The compounds can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formula-tions for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, with an added preservative. The compositions can take such forms as suspensions, solutions, or emul-sions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.
Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Addition-ally, suspensions of the active compounds can be prepared as appropriate oily injection suspensions.
Suitable lipophilic solvents or vehicles include fatty oils or synthetic fatty acid esters. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension. Option-ally, the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds and allow for the preparation of highly concentrated solutions. Alternatively, a present composition can be in powder form for con-stitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
Compounds of the present invention also can be formulated in rectal compositions, such as suppositories or retention enemas, e.g., containing conventional suppository bases. In addition to the formulations described previously, the compounds also can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (for example, subcutaneously or intra-muscularly) or by intramuscular injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble deriva tives, for example, as a sparingly soluble salt.
Many of the compounds of the present invention can be provided as salts with pharmaceuti cally compatible counterions. Such pharmaceutically acceptable base addition salts are those salts that retain the biological effectiveness and properties of the free acids, and that are obtained by reaction with suitable inorganic or organic bases.
In particular, a compound of formula (I) can be administered orally, buccally, or sublin-gually in the form of tablets containing excipients, such as starch or lactose, or in capsules or ovules, either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents. Such liquid prepara-tions can be prepared with pharmaceutically accept-able additives, such as suspending agents. A com-pound also can be injected parenterally, for ex-ample, intravenously, intramuscularly, subcutane-ously, or intracoronarily. For parenteral admin-istration, the compound is best used in the form of a sterile aqueous solution which can contain other substances, for example, salts, or monosaccharides, such as mannitol or glucose, to make the solution isotonic with blood.
For veterinary use, a compound of formula (I) or a nontoxic salt thereof, is administered as a suitably acceptable formulation in accordance with normal veterinary practice. The veterinarian can readily determine the dosing regimen and route of administration that is most appropriate for a particular animal.
Thus, the invention provides in a further aspect a pharmaceutical composition comprising a compound of the formula (I), together with a pharma-ceutically acceptable diluent or carrier therefor.
There is further provided by the present invention a process of preparing a pharmaceutical composition comprising a compound of formula (I), which process comprises mixing a compound of formula (I), together with a pharmaceutically acceptable diluent or carrier therefor.
In a particular embodiment, the invention includes a pharmaceutical composition for the cura-tive or prophylactic treatment of erectile dysfunc-tion in a male animal, or arousal disorder in a female animal, including humans, comprising a com-pound of formula (I) or a pharmaceutically accept-able salt thereof, together with a pharmaceutically acceptable diluent or carrier.
Compounds of formula (I) can be prepared by any suitable method known in the art, or by the following processes which form part of the present invention. In the methods below, R°, R1, R2, R3, and R4, as well as Ra through Rh, Q, B, C, D, E, X, and Y, are defined as in structural formula (I) above.

In particular, compounds of structural formula (I) can be prepared according to the following synthetic scheme.
Daugan U.S. Patent No. 5,859,006, in-s corporated herein by reference, discloses prepara-tion of a compound of structural formula (III):
O
COAlkyl ~RO~ q N
N H
H

(III) The compounds of structural formula (I) can be pre-pared from compounds similar to the compound of structural formula (III) using appropriate RZ and R4 substituents.
The following illustrates a general method of synthesizing a compound of structural formula (I) .

Ro 0 ,,~~~ iCH3 ~' 0 LiOH
N
H H _ H-Cl OJ
(IV) Ro 0 \ ..,1~
~' OH 1) triphosgene N
H H = H-Cl 2 ) RaRb_~

O
(V) Ro 0 ,Ra N
' cinnamic acid, N NH Rb N-methylmorpholine H H
Cl N~ O~
/ ~ ~ CH3 N /N
O

(VI) Ra~N,Rb ,,~~ 0 /
N \ \
N HY a a H

O
(VII) Known compound (IV) (see Daugan U.S.
Patent No. 5,859,006) is hydrolyzed to carboxylic acid (V), which then is treated with triphosgene followed by treatment with a desired amine to pro-vide amide (VI). Amide (VI) then is treated with a carboxylic acid ir~ the presence of an activating group, or other carboxylic acid derivative (e. g., acid chloride), having the desired -(X)n-Y residue and a base to provide bisamide (VII). Bisamide (VII) is a compound of structural formula (I).
The following illustrates a synthesis of compounds (V)-(VII) from compound (IV), wherein R° is hydrogen. The synthesis of compound (IV) can be found in Daugan U.S. Patent No. 5,859,006.

iCH3 1~ 0 LiOH

H = H-Cl \

(IVa) (IV, wherein R°=H) O
- OH

1. triphosgene H = H-C1 2. diethylamine O--l (Va) (v, wherein R°=H) ,.~~~ iCH3 1. N
\CH3 N
H = cinnamic acid, N-methylmorpholine Cl ~ N~ 0~
0 ~ ~ CH3 0~ N / N .
(VIa) 0 CH3~
(VI, wherein R°=H
and Ra and Rb=CH3) CH3~N,CH3 /\
~. O
N~N
H
O
O
(VIIa) (VII, wherein R°=H and Ra and Rb=CH3) Preparation of (1R,3R)-1-benzo[1,3)dioxol-5-yl-2,3,4,9-tetrahydro-1H-(3-carboline-3-carboxylic acid hydrochloride (Va) Lithium hydroxide (0.32 g, 13.2 mmol) was added to a stirred mixture of (1R,3R)-1-benzo[1,3]-dioxol-5-yl-2,3,4,9-tetrahydro-1H-(3-carboline-3-carboxylic acid methyl ester (IVa) (2.5 g, 6.5 mmol) in 10 mL of anhydrous tetrahydrofuran at room temp-erature. Methanol (1 mL) was added to help dissolu-tion of the solids. After stirring overnight at room temperature, the reaction mixture was parti-tioned between ethyl acetate (10 mL) and deionized water (25 mL). The layers were separated, and the pH of the aqueous layer was adjusted to 2.3 with 1 M
HC1. After 1 hour, the resulting slurry was filtered, then the filter cake was washed with a quantity of deionized water sufficient to remove the yellow color. The resulting solid was dried under vacuum to give 1.38 g (57%) of compound (Va) as an off-white solid.
1H NMR (DMSO-d6) ~: 10.70 (s, 1H) , 7.52 (d, J=7 Hz, 1H), 7.28 (d, J=8 Hz, 1H), 7.13-7.01 (m, 5H), 6.07 (s, 2H), 5.78 (s, 1H), 4.22 (dd, J=5, 11 Hz, 1H), 3.38 (dd, J=5, 15 Hz, 1H), 3.21 (dd, J=2, 15 Hz, 1H); MS ES+m/e 337 (p+1), ES-m/e 335 (p-1); IR (KBr, cm~l) 3621, 3450, 1758.
Preparation of (1R,3R)-1-benzo[1,3]dioxol-5-yl-2-(3-phenyl-acryloyl)-2,3,4,9-tetrahydro-1H-~i-carboline-3-carboxylic acid dimethylamide (VIa) Triethylamine (1.5 mL, 10.7 mmol) was added to a stirred mixture of compound (Va) (2 g, 5.4 mmol) and 60 mL of anhydrous tetrahydrofuran at room tempterature. Triphosgene (1.6 g, 5.4 mmol) was added to the slurry, then stirring was continued for 2 hours at room temperature. Dimethylamine was condensed into a dropping funnel (about 1 mL), and was added directly to the stirred slurry. After 1 hour, the reaction mixture was filtered and the volatiles were removed from the filtrate under vacuum. The resulting foam was subjected to flash chromatography (silica, EtOAc) to give a 1.65 g (86 0 ) of compound (VIa) as a yellow foam.
1H NMR (DMSO-d6) ~: 10.38 (s, 1H), 7.44 (d, J=6 Hz, 1H), 7.21 (d, J=7 Hz, 1H), 7.03-6.88 (m, 3H), 6.83-6.79 (m, 2H), 5.99 (s, 2H), 5.13 (d, J=7 Hz, 1H), 4.04 (m, 1H), 3.12 (s, 3H), 2.86 (s, 3H), 2.8-2.76 (m, 2H); MS ES+m/e 364 (p+1), ES-m/e 362 (p-1); IR
(KBr, cm-1) 3463, 1642.
Preparation of (1R,3R)-1-benzo[1,3]dioxol-5-yl-2-(3-phenylacryloyl)-2,3,4,9-tetrahydro-1H-(3-carboline-3-carboxylic acid dimethylamide (VIIa) 2-Chloro-3,5-dimethyoxy-2,4,6-triazine (0.5 g, 2.8 mmol) was added to a mixture of trans-cinnamic acid in 15 mL of anhydrous tetrahydrofuran.
N-Methylmorpholine (0.32 mL, 2.9 mmol) was added and the resulting solution was stirred at room tempera-ture for 1.75 hours. Compound (VIa) was added, then the reaction was stirred for 4 hours at room temper-ature. The reaction mixture was partitioned between ethyl acetate (25 mL) and 1 M HCl (25 mL). The layers were separated, and the organic layer was washed with 25 mL of saturated sodium bicarbonate solution followed by 25 mL of brine. The EtOAc was dried over MgS04, filtered, and removed under vacuum to give the crude product as a yellow foam. The product was purified by flash chromatography (50%
hexane, 50% CHzCl2 to CHzCl2 to EtOAc) to yield 0.613 g (42 0) of compound (VIIa) as a yellow foam: mp 175-178°C.
1H NMR (DMSO-d6) ~: 10.82, 10.75 (overlapping br s, 1H), 7.89-7.21 (m, 9H), 7.11-6.67 (m, 5H), 6.65 (br s, 1H), 5.92 (s, 2H), 5.70-5.63 (m, 1H), 2.9 (dd, J=6, 15 Hz, 1H), 2.75 (br s, 1H), 2.58 (br s, 2H), 2.51-2.48 (m, 1H), 2.36 (br s, 2H), 2.18 (br s, 1H);
MS ES-m/e 492 (p-1).
The following scheme illustrates the synthesis of a compound of structural formula (I) from ester (IV), and illustrates an alternative method of synthesizing a bisamide (VII).

..,1~
OMe / N~~~ cinnamic acid - DCC, Et3N
/

(IVa) \ 'w~ /
OMe / N~~N \ \
H = 0 \

(VIII) O
\ ~~~1~ /
NMe2 / N~~ \ \
AlMe3/Me2HN~HC1 H H~ O
Example 1 O
O
The above scheme illustrates the synthesis of a compound of structural formula (II), i.e., compound (VIII), directly from compound (IVa) by reaction with a carboxylic acid having the desired (X)n-Y
residue, in the presence of dicyclohexylcarbodiimide and triethylamine. Compound (VIII) then can be converted to another compound of structural formula (I), i.e., Example 1, by a reaction using trimethyl aluminum and dimethylamine hydrochloride. The reaction between compound (IVa) and cinnamic acid is set forth in Bombrun U.S. Patent No. 6,117,881, incorporated in its entirety herein by reference.
It should be understood that protecting groups can be utilized in accordance with general principles of synthetic organic chemistry to provide compounds of structural formula (I). Protecting group-forming reagents, like benzyl chloroformate and trichloroethyl chloroformate, are well known to persons skilled in the art, for example, see T.W.
Greene et al., "Protective Groups in Organic Synthe-sis, Third Edition," John 4diley and Sons, Inc., NY, NY (1999). These protecting groups are removed when necessary by appropriate basic, acidic, or hydro-genolyt.ic conditions known to persons skilled in the art. Accordingly, compounds of structural formula (I) not specifically exemplified herein can be pre-pared by persons skilled in the art.
In addition, compounds of formula (I) can be converted to other compounds of formula (I).
Thus, for example, a particular R substituent can be interconverted to prepare another suitably substi-tuted compound of formula (I). Examples of appro-priate interconversions include, but are not limited to, ORa to hydroxy by suitable means (e.g., using a agent such as BBr3, or a palladium catalyst, like palladium-on-carbon, and hydrogen), or amino to substituted amino, such as acylamino or sulphonyl-amino, using standard acylating or sulfonylating conditions.
Compounds of formula (I) can be prepared by the method above as individual stereoisomers or as a racemic mixture. Individual stereoisomers of the compounds of the invention can be prepared from racemates by resolution using methods known in the art for the separation of racemic mixtures into their constituent stereoisomers, for example, using HPLC on a chiral column, such as Hypersil naphthyl urea, or using separation of salts of stereoisomers.
Compounds of the invention can be isolated in association with solvent molecules by crystalliza-tion from, or evaporation of, an appropriate sol-vent.
The pharmaceutically acceptable acid addi-tion salts of the compounds of formula (I) that con-tain a basic center can be prepared in a convention-al manner. For example, a solution of the free base can be treated with a suitable acid, either neat or in a suitable solution, and the resulting salt isolated either by filtration or by evaporation under vacuum of the reaction solvent. Pharmaceut-ically acceptable base addition salts can be ob-tamed in an analogous manner by treating a solution of a compound of formula (I) with a suitable base.
Both types of salt can be formed or interconverted using ion-exchange resin techniques. Thus, accord-ing to a further aspect of the invention, a method for preparing a compound of formula (I) or a salt or solvate (e.g., hydrate) is provided, followed by (i) salt formation, or (ii) solvate (e. g., hydrate) formation.
The following additional abbreviations are used hereafter in the accompanying examples: rt (room temperature), min (minute), h (hour), g (gram), mmol (millimole), m.p. (melting point), LiOH
(lithium hydroxide), eq (equivalents), L (liter), mL

(milliliter), ,uL (microliter), DMSO (dimethyl sul-foxide), Et3N (triethylamine), MeNH2 (methylamine), THF (tetrahydrofuran), Me2NH (dimethylamine), DCC
(1,3-dicyclohexylcarbodiimide), AlMe3 (trimethyl-aluminum), Me (methyl), EtOAc (ethyl acetate), CHC13 (chloroform) , and Na2S04 (sodium sulfate) .
Example 2 S
~ ..,1~ /
/ N
I H~' H ~ O
O
Example 2 was prepared from Compound (IVa) by the following synthetic sequence.
AlMe3/Me2HN~HC1 Compound (IVa) O
...1~
NMe2 / NH Lawesson's N/~ Reagent H _ H /
O
O

S
...1~
\ ~, NMe2 / NH
N/ H'= Cinnamic acid H - Example 2 / DCC, Et3N
\
O
O
Example 3 (-) - (1R, 3R) -1-Benzo [1, 3] dioxol-5-yl-1, 3, 4, 9-tetra-hydro-beta-carboline-2,3-dicarboxylic acid 3-methyl ester 2-(4-nitrophenyl)ester H
...1~ ~CH3 N ~ /
H H
/ O \ N02 Example 3 was prepared from Compound (IVa) as follows. Also see F. Pinnen et al., J. Chem.
Soc., Perkin Trans. I, 12, p. 1611 (1994) .

Et3N, THF, 0°C
Compound (IVa) Example 3 O
C1 O.
78%
4-Nitrophenyl chloroformate was added dropwise to a suspension of Compound (IVa) (2.0 g, 52 mmol ) and Et3N ( 1 . 8 mL, 13 mmol ) in THF ( 50 mL) at 0°C under a nitrogen blanket. The resulting mixture was slowly warmed to room temperature and stirred for 18 hours, then diluted with EtOAc (200 mL). The resulting solution was washed with brine (100 mL), dried over Na2S04, filtered, and the solvents were removed under reduced pressure. The residue was purified by flash column chromatography, eluting with EtOAc/CHC13 (1:19) to provide Example 3 as a yellow powder (2.08 g, 78%): mp 175-187°C; TLC Rf ( 9 : 1 CHC13/EtOAc ) =0 . 84 .
1H NMR (300 MHz, DMSO-ds) b: 10.90 (s, 1H), 8.33 (d, J=9.0 Hz, 2H), 7.58-7.56 (m, 3H), 7.30 (d, J=7.7 Hz, 1H), 7.07 (dt, J=6.5, 21.8 Hz, 2H), 6.88 (bs, 1H), 6.72 (s, 1H), 6.58 (bs, 1H), 6.44 (bs, 1H), 6.01 (s, 2H), 5.70 (bs, 1H), 5.45 (bs, 1H), 3.40 (d, J=13.7 Hz, 1H), 3.28-3.21 (m, 1H); API MS m/z 516 [C2.,L.i21N30a+H~ ~; (a~ D25°C_-121 . 7° (c=1 . 0, DMSO) . Anal .
Calcd. for Cz.,H21N308: C, 62.91; H, 4.11; N, 8.15.
Found: C, 62.54; H, 3.91; N, 8.02.
The following Examples 4-12 were prepared in a manner similar to Examples 1-3.

O~O~CH3 w H H ~/ II
to Example 4 H ~ J

O
~CH3 Example 5 O
O
to Example 6 H
O~CH3 Example 7 Preparation of Example 8 O
H
,,. ~ ~CH3 N
H
CH
N ~ ~ 3 H H/=

\

Example 8 Example 8 was prepared by the following synthetic sequence O
/ \ ., H
N~NH triethylamine 1) triphosgene H _ Example 8 (Va) 2) propylamine - acetyl chloride O
O-(Ix) Preparation of (1R)-(+)-1-benzo[1,3]dioxol-5-yl-2,3,4,9-tetrahydro-1H-~i-carboline-3-carboxylic acid propylamide (Compound (IX)) A suspension of compound (Va) (2.0 g, 6.0 mmol) and triethylamine (2 mL) in tetrahydrofuran (50 mL) was immersed in a sonicator until all solids dissolved. Triphosgene (1.6 g, 5.4 mmol) was added, and the resulting suspension was stirred for 6 hours without cooling. A colorless solid was removed by filtration, then the filtrate was concentrated to a foam. The foam was dissolved in chloroform (50 mL) and n-propylamine (2 mL) was added. The reaction was stirred for 18 hours without cooling, the solvent was evaporated, and the residue purified by chromatography (silica gel, 75% ethyl acetate: 25s hexanes) to give 1.4 g (61.%) of compound (IX) as a solid.
1H NMR (DMSO-d6) d: 10.71 (s, 1H) , 7.84 (t, J=8 Hz, 1H), 7.45-6.59 (m, 7H), 6.0 (s, 2H), 5.15 (s, 1H), 3.45 (m, 1H), 3.04 (q, J=8 Hz, 2H), 2.8 (dd, J=5, 13 Hz, 1H), 1.4 (m, 2H), 0.84 (t, J=8 Hz, 3H); MS
ES+m/e 378.2 (p+1), ES-m/e 376.2 (p-1); Anal. Calcd.
for C22H2aNsOa; C, 70.01; H, 6.14; N, 11.13. Found:
C, 70.30; H, 6.23; N, 11.27.
Preparation of (1R)-(+)-2-acetyl-1-benzo[1,3]dioxol-5-yl-2,3,4,9-tetrahydro-1H-(3-carboline-3-carboxylic acid propylamide (Example 8) To a solution of compound (IX) (1.4 g, 3.7 mmol) and triethylamine (2 mL) in methylene chloride (25 mL) was added acetyl chloride (0.36 mL, 5.0 mmol), then the mixture was stirred for 18 hours at room temperature. The solution was washed once with water, once with saturated sodium chloride solution, dried (sodium sulfate), filtered, and concentrated in vacuo to provide a solid that was recrystallized (ethyl acetate) to give 310 mg (200) of Example 8 as a solid: mp 163-165°C.
1H NMR (DMSO-d6) d: 10.88 (s, 1H) , 7.95 (t, J=5 Hz, 0.6H (rotamer)), 7.7 (t, J=5 Hz, 0.4H (rotamer)), 7.4-6.7 (m, 7H), 5.85-6.2 (m, 3H), 5.25 (br s, 0.6H
(rotamer)), 4.95 (br s, 0.4 H (rotamer)), 3.35 (dd, J=6, 13 Hz, 2H), 2.85 (m, 2H), 2.0 (s, 1.8H
(rotamer)), 1.9 (s, 1.2H (rotamer)), 1.2 (m, 2H), 0.62 (m, 3H); MS ES+m/e 420.2 (p+1), ES-m/e 418.3 (p-1) ; Anal. Calcd. for C24H25N3~4~ C. 68.71; H, 6.01;
N, 10.02. Found: C, 68.57; H, 5.69; N, 10.02.
Preparation of Example 9 O
N
/ \
o CH
H ~ 3 HY

O
Example 9 Example 9 was prepared by the following synthetic sequence:

/\
- i ' N'1 N~NH ~O
1) triphosgene (Va) H - triethylamine 2) morpholine / Example. 9 acetyl chloride O
O-J
(X) Preparation of (1R)-(+)-(1-benzo[1,3]dioxol-5-yl-2,3,4,9-tetrahydro-1H-~i-carbolin-3-yl)-morpholine-4-yl-methanone (X) A suspension of compound (Va) (350 mg, 1.04 mmol) and triethylamine (0.5 mL) in tetra-hydrofuran (10 mL) was sonicated until all solids were dissolved. Triphosgene (300 mg, 1.0 mmol) was added and the resulting suspension was stirred for 2 hours. A colorless solid was removed by filtration and the filtrate was concentrated in vacuo to pro-vide a foam. The foam was dissolved in methylene chloride (15 mL), treated with morpholine (0.5 mL), and the resulting solution stirred for 18 hours.
The solvent was evaporated in vacuo, and the crude material was purified by chromatography (silica gel, ethyl acetate) to give 280 mg (660) of compound (X) as a solid.
1H NMR (DMSO-d6) b: 10.35 (s, 1H), 7.45-6.77 (m, 7H), 6.02 (s, 2H), 5.15 (s, 1H), 4.06 (dd, J=4, 13 Hz), 3.7-3.45 (m, 8H), 2.85 (dd, J=4, 13 Hz, 2H), 2.1 (t, J=10 Hz, 1H); MS ES+m/e 406.0 (p+1), ES-m/e 404.2 (p-1) .
Preparation of (1R) - (+) -1- [benzo [1, 3] dioxol-5-yl-3-(morpholine-4-carbonyl)-1,3,4,9-tetrahydro-(3-carbolin-2-yl]-ethanone (Example 11) To a solution of compound (X) (1.7 g, 4.2 mmol) and triethylamine (2 mL) in methylene chloride (50 mL) was added acetyl chloride (3.29 mg, 4.2 mmol, 0.3 mL). The reaction was stirred for 18 hours without cooling. An additional quantity of acetyl chloride (0.1 mL) was added, and stirring was continued for 30 minutes. The reaction was concentrated and purified by chromatography (silica gel, 75o ethyl acetate: 25% hexanes, then ethyl acetate) to give 800 mg (420) of Example 9 as a colorless solid.
1H NMR (DMSO-d6) b: 10.8 (s, 1H), 7.55-6.8 (m, 7H), 6.75 (br s, 0.6H (rotamer)), 6.62 (br s, 0.4H
(rotamer)), 5.97 (d, J=4 Hz, 2H), 5.6 (br s, 0.6H
(rotamer)), 5.75 (br s, 0.4H (rotamer)), 3.54-2.95 (m, 9H) , 2 .85 (dd, J=5, 13 Hz, 1H) , 2 .3 (s, 3H) ; MS
ES+m/e 448.1 (p+1), ES-m/e 446.2 (p-1); Anal. Calcd.
for C25HzsNaOs : C, 67 . 10 ; H, 5 . 63 ; N, 9 . 3 9 . Found C, 67.00; H, 5.69; N, 9.28.

O ~N~CH3 ~N~
\ /CH3 NN
O
N H ' H / O
i O

Example 10 Preparation of Example 11 O

H i N
H H

Example 11 Example 11 was prepared using the following synthetic sequence:

H
N
H
N
n-butylamine H
(Va) _ H _ O
O
(XI) trans-cinnamic acid N-methylmorpholine Example 11 C1 N~ OCH3 N /N

Preparation of (1R,3R)-(+)-1-benzo[1,3]dioxol-5-yl 2,3,4,9-tetrahydro-1H-(3-carboline-3-carboxylic acid butylamide (XI) To a solution of compound (Va) (2.17 g, 6 mmol) in chloroform (50 mL) was added n-butylamine (585 mg, 6 mmol). The reaction was stirred for 18 hours at room temperature. The solvent was con-centrated in vacuo, then the residue was purified by chromatography (silica gel, 50% ethyl acetate: 70%
hexanes) to give 1.7 g (72%) of compound (XI) as a colorless foam.
1H NMR (DMSO-d6) ~: 10.28 (s, 1H) , 7.6 (t, J=6 Hz, 1H), 7.45-6.85 (m, 8H), 6.0 (s, 2H), 5.08 (d, J=7 I Hz, 1H), 3.55 (m, 1H), 3..14 (q, J=6 Hz, 2H), 2.97 (dd, J=3, 16 Hz, 1H), 2.7 (dt, J=3,16 Hz), 1.47 (m, 2H), 1.3 (m, 2H), 0.82 (t, J=6 Hz, 3H); MS ES+m/e 392.3 (p+1), ES-m/e 390.4 (p-1); Anal. Calcd. for 1O C23HZ,N3O3: C, 70.57; H, 6.43; N, 10.73. Found: C, 70.34; H, 6.11; N, 10.53.
Preparation of (1R, 3R) - (+) -1-benzo [1, 3] dioxol-5-yl-2-(3-phenylacryloyl)-2,3,4,9-tetrahydro-1H-(3-carboline-3-carboxylic acid butylamide (Example 11) A mixture of trans-cinnamic acid (376 mg.
2.5 mmol), 2-chloro-3,5-dimethoxy-2,4,6-triazine (500 mg, 2.8 mmol), N-methylmorpholine (0.32 mL, 2.9 mmol), and tetrahydrofuran (20 mL) was stirred without cooling for 1.5 hours. Compound (XI) (1.0 g, 2.5 mmol) was added, then the reaction was stirred for an additional 18 hours. The solvent was evaporated, and the residue was dissolved in ethyl acetate, washed once with 1N hydrochloric acid, once with saturated sodium bicarbonate solution, dried (sodium sulfate), filtered, and concentrated in vacuo. Chromatography (silica gel, 30% ethyl acetate: 70% hexanes) of the residue provided 800 mg (61a) of Example 11 as a solid: mp 114-117°C.
1H NMR (DMSO-d6) b: 10.8 (s, 1H), 6.8-7.7 (m, 14H), 5.95 (s, 2H), 5.3 (m, 1H), 3.0 (m, 1H), 2.78 (m, 1H), 1.15 (m, 6H), 0.8 (m, 3H); MS ES+m/e 522.02 (p+1) , ES-m/e 520.13 (p-1) , IR (KBr, cm-1) 1674, 1644, 1503, 1487; Anal. Calcd. for C32H31N304: C, 73.68; H, 5.99; N, 8.06. Found: C, 73.33; H, 5.96;
N, 8.25.
Preparation of Example 12 ~,CH3 ~ °
N
H H

Example 12 Example 12 was prepared using the following synthetic sequence:

...1~
NH

H -(Va) 1) triphosgene /
2) methylamine 0-l (XII) - Example 12 trans-cinnamic acid, N-methylmorpholine C1 N~ O~

N~N
IYO
CH3~
Preparation of (1R,3R)-1-benzo[1,3]dioxol-5-yl-2-(3-phenylacryloyl)-2,3,4,9-tetrahydro-1H-~i-carboline-3-carboxylic acid methylamide (XII) Triethylamine (2 mL, 14 mm, 2.4 eq) was added to a stirred mixture of compound (Va) (2 g, 5.2 mmol) and anhydrous tetrahydrofuran (25 mL) at room temperature. The resulting mixture was soni-cated until complete dissolution was achieved.

Triphosgene (1.6 g, 5.4 mmol) was added to the slurry, and stirring was continued for 5 hours at room temperature. The solids were removed by filtration and the filtrate concentrated in vacuo.
The residue was dissolved in chloroform, and a solution of 2 M methylamine in tetrahydrofuran (10 mL, 20 mmol) was added. After 1.5 hours, the reaction mixture was diluted with water and shaken.
The organic layer was dried (sodium sulfate), filtered, and concentrated in vacuo. The residue (compound (XII) was used as is.
1H NMR (DMSO-d6) ~: 10.38 (s, 1H), 7.44 (d, J=6 Hz, 1H), 7.21 (d, J=7 Hz, 1H), 7.03-6.88 (m, 3H), 6.83-6.79 (m, 2H), 5.99 (s, 2H), 5.10 (m, 1H), 3.5-3.6 (m, 1H), 3.30 (s, 3H), 3.00 (dd, J=1.5, 11.4 Hz, 1H), 2.72 (dd, J=2.5, 11.3 Hz, 1H), 2.64 (s, 3H), 2.49 (s, 3H); MS ES+m/e 350.2 (p+1), ES-348.2 (p-1).
Preparation of (1R,3R)-~2-[(2E)-3-(phenyl)prop-2-enoyl]-1-(2H-benzo(d]1,3-dioxolan-5-yl-(1,2,3,4-tetrahydrobetacarbolin-3-yl)~-N-methylcarboxamide (Example 12) 2-Chloro-3,5-dimethoxy-2,4,6-triazine (0.5 g, 2.8 mmol) was added to a solution of trans-cinnamic acid (0.376 g, 2.5 mmol) in anhydrous tetrahydrofuran (20 mL). N-Methylmorpholine (0.32 mL, 2.9 mmol) was added, then the resulting solution was stirred at room temperature for 1.5 hours. A
solution of compound (XII) (0.873 g, 2.50 mmol) in anhydrous tetrahydrofuran (5 mL) was added, and the reaction was stirred for 2 hours at room tempera-ture. The reaction mixture was partitioned between ethyl acetate and 1M hydrochloric acid. The layers were separated, and the organic layer was washed once with saturated sodium bicarbonate solution and once with saturated sodium chloride solution. The organic layer was dried (sodium sulfate), filtered, and concentrated in vacuo to give the crude product as a yellow foam. The product was purified by flash chromatography (silica gel, 50o hexane/50o ethyl acetate to ethyl acetate) to provide 0.43 g (35%) of Example 12 as a yellow foam. MS FAB exact mass calculated for C29H23N4O4: m/z=479.1845. Found:
479.1845.
Preparation of Example 13 CH3 ~NH

_ ~~ O
N \ \
H H- _ \

O
Example 13 Example 13 was prepared in an identical manner to Example 12 and substituting p-chloro trans cinnamic acid for trans cinnamic acid.
1H NMR (DMSO-d6) b : 2 . 18 (br s, 1 . 5H) , 2 . 35 (br S, 1H), 2.50 (m, 1H), 2.51 (br s, 2H), 2.73 (br s, 1H), 2.77 (dd, J=5.8 Hz, 15 Hz, 1H), 3.24-3.40 (m, 0.5H), 5.75 (br s, 1H), 5.95 (s, 2H), 6.68 (br s, 1H), 6.78-7.10 (m " 5H), 7.26-7.37 (m, 1H), 7.46-7.50 (m, 7H), 10.75 (br s, 0.5H), 10.88 (br s, 0.5H); MS
ES+m/e 528.2 (p+1), MS ES-m/e 526.2 (p-1).
The following intermediates have been prepared, and can be used to provide a composition of structural formula (I) by the methods set forth above. In particular, an intermediate is reacted with a carboxylic acid, or a carboxylic acid deriva-tive, having the desired (X)n-Y residue. As demon-strated hereafter, the following intermediates also are potent and selective inhibitors of PDE5, and, like compounds of structural formula (I), can be used in a variety of therapeutic areas wherein such inhibition is considered beneficial.
O ~O~CH3 NH
\ N H %/
H
\ ~ iCH3 Intermediate la to Intermediate 1b H
O~CH3 ~CH3 Intermediate 2 O vO~CH3 / NH
\

\ N H ~/
H
\
Intermediate 3 n n l! 1 Intermediate 4 H
~CH3 Intermediate 5 H
0 ~N

Intermediate 6 O~CH3 Intermediate 7a Intermediate 7b _ 75 H /
O ~N \

Intermediate 8 Intermediate 9a 2 5 O ~O~CH3 / NH
\ N H ~/ O

\ O
Intermediate 9b H

Intermediate 10 Intermediate 11 ~0 H
0 ~N NJ

Intermediate 12 _ 77 _ ~ ~N
H
O ~N \
O
O
Intermediate 13a N
H ~ J

Intermediate 13b _ 78 _ H
~F

Intermediate 14 H

Intermediate 15a H
Intermediate 15b i ~CH3 H
o~CH3 Intermediate 16a ~CH3 H
O~N \ O~CH3 NH

N H ,/
H
\ 0 Intermediate 16b ~CH3 ~CH3 25 Intermediate 17a O
H ~ ( ~CH3 OiCH3 Intermediate 17b H
f~
~0 Intermediate 18 H

~o>
Intermediate 19 Intermediate 20a J,~>

Intermediate 20b J

Intermediate 21 H
/N~ N

Intermediate 22 H
Intermediate 23 H
O

Intermediate 24a H

n NH
~ ~ H~ ~ O
'N
H
Intermediate 24b H

Intermediate 25 H
25 Intermediate 26a _ 87 _ H
O ~N

Intermediate 26b F
H
O~N
2 0 /~ ~( NH
N H ::/ 0 H
O
Intermediate 27 _ 88 H

Intermediate 28 H
i Intermediate 29a H
0'\ /N O~

NH
/
i ~ ' O
\ N H %/
H
\ 0 Intermediate 29b H ~ F
/

Intermediate 30a \ F
H
Intermediate 30b H
O N N
O
/~ ,( NH
\ :/ 0 N H
H \ 0 Intermediate 31 NHZ
\
H
25 Intermediate 32 O
H ~ ~ ~CH3 \ OiCH3 ~CH3 O
Intermediate 33 O
H ~ ~ ~CH3 \ OiCH3 Intermediate 34 H

H

O
O.
Intermediate 35 H ~ ~~CH3 iCH3 -O
~CH3 Intermediate 36 H

~CH3 \ O~CH3 Intermediate 37 H

~CH3 25 Intermediate 38 Intermediate 39 Compounds of the present invention can be formulated into tablets for oral administratlOTl.
For example, a compound of formula (I) can be formed into a dispersion with a polymeric carrier by the coprecipitation method set forth in WO 96/38131, incorporated herein by reference. The coprecipi-tated dispersion then can be blended with excipi-ents, then pressed into tablets, which optionally are film-coated.
The compounds of structural formula (I) were tested for an ability to inhibit PDE5. The ability of a compound to inhibit PDES activity is related to the ICSO value for the compound, i.e., the concentration of inhibitor required for 50o inhibi-tion of enzyme activity. The ICso value for com-pounds of structural formula (I) were determined using recombinant human PDE5.
The compounds of the present invention typically exhibit an ICSO value against recombinant human PDE5 of less than about 50 ,uM, and preferably I

less than about 25 ,uM, and more preferably less than about 15 ,um. The compounds of the present invention typically exhibit an ICso value against recombinant human PDE5 of less than about 1 ,uM, and often less than about 0.5 ,uM. To achieve the full advantage of the present invention, a present PDE5 inhibitor has an ICS of about 0.1 nM to about 15 ,uM.
The production of recombinant human PDEs and the ICso determinations can be accomplished by well-known methods in the art. Exemplary methods are described as follows:
EXPRESSION OF HUMAN PDEs Expression in Saccharomyces cerevisiae (Yeast) Recombinant production of human PDE1B, PDE2, PDE4A, PDE4B, PDE4C, PDE4D, PDE5, and PDE7 was carried out similarly to that described in Example 7 of U.S. Patent No. 5,702,936, incorporated herein by reference, except that the yeast transformation vector employed, which is derived from the basic ADH2 plasmid described in Price et al., Methods in Enzymology, 185, pp. 308-318 (1990), incorporated yeast ADH2 promoter and terminator sequences and the Saccharomyces cerevisiae host was the protease-defi-cient strain BJ2-54 deposited on August 31, 1998 with the American Type Culture Collection, Manassas, Virginia, under accession number ATCC 74465. Trans-formed host cells were grown in 2X SC-leu medium, pH
6.2, with trace metals, and vitamins. After 24 hours, YEP medium-containing glycerol was added to a final concentration of 2X YET/3% glycerol. Approxi-mately 24 hr later, cells were harvested, washed, and stored at -70°C.
HUMAN PHOSPHODIESTERASE PREPARATIONS
Phosphodiesterase Activity Determinations Phosphodiesterase activity of the prepara-tions was determined as follows. PDE assays utiliz-ing a charcoal separation technique were performed essentially as described in Loughney et al. (1996).
In this assay, PDE activity converts [32P]CAMP or [32P] cGMP to the corresponding [32P] 5' -AMP or [32P]5'-GMP in proportion to the amount of PDE ac-tivity present. The [32P]5'-AMP or [32P]5'-GMP then was quantitatively converted to free [32P]phosphate and unlabeled adenosine or guanosine by the action of snake venom 5'-nucleotidase. Hence, the amount of [32P]phosphate liberated is proportional to en-zyme activity. The assay was performed at 30°C in a 100 ,uL reaction mixture containing (final concentra-tions) 40 mM Tris HCl (pH 8.0) , 1 ,uM ZnS04, 5 mM
MgCl2, and 0.1 mg/mL bovine serum albumin (BSA). PDE
enzyme was present in quantities that yield <300 total hydrolysis of substrate (linear assay condi-tions). The assay was initiated by addition of substrate (1 mM [32P]CAMP or cGMP), and the mixture was incubated for 12 minutes . Seventy-five ( 75 ) ,ug of Crotalus atrox venom then was added, and the incubation was continued for 3 minutes (15 minutes.
total). The reaction was stopped by addition of 200 ,uL of activated charcoal (25 mg/mL suspension in 0.1 M NaH2P04, pH 4). After centrifugation (750 X g for 3 minutes) to sediment the charcoal, a sample of the supernatant was taken for radioactivity determina-tion in a scintillation counter and the PDE activity was calculated.
Purification of PDE5 from S. cerevisiae Cell pellets (29 g) were thawed on ice with an equal volume of Lysis Buffer (25 mM Tris HCl, pH 8, 5 mM MgCl2, 0.25 mM DTT, 1 mM benzamidine, and 10 ,uM ZnS04). Cells were lysed in a Microfluid-izer (Microfluidics Corp.) using nitrogen at 20,000 psi. The lysate was centrifuged and filtered through 0.45 ,um disposable filters. The filtrate was applied to a 150 mL column of Q SEPHAROSE Fast-Flow (Pharmacia). The column was washed with 1.5 volumes of Buffer A (20 mM Bis-Tris Propane, pH 6.8, 1 mM MgClz, 0.25 mM DTT, 10 ,uM ZnS04) and eluted with a step gradient of 125 mM NaCl in Buffer A followed by a linear gradient of 125-1000 mM NaCl in Buffer A. Active fractions from the linear gradient were applied to a 180 mL hydroxyapatite column in Buffer B (20 mM Bis-Tris Propane (pH 6.8), 1 mM MgCl2, 0.25 mM DTT, 10 ,uM ZnS04, and 250 mM KC1). After load-ing, the column was washed with 2 volumes of Buffer B and eluted with a linear gradient of 0-125 mM
potassium phosphate in Buffer B. Active fractions were pooled, precipitated with 60% ammonium sulfate, and resuspended in Buffer C (20 mM Bis-Tris Propane, pH 6.8, 125 mM NaCl, 0.5 mM DTT, and 10 ,uM ZnS04).
The pool was applied to a 140 mL column of SEPHACRYL S-300 HR and eluted with Buffer C.

_ 98 -Active fractions were diluted to 50o glycerol and stored at -20°C.
The resultant preparations were about 85%
pure by SDS-PAGE. These preparations had specific activities of about 3 umol cGMP hydrolyzed per min-ute per milligram protein.
Inhibitory Effect on cGMP-.PDE
cGMP-PDE activity of compounds of the present invention was measured using a one-step assay adapted from Wells et al., Biochim. Biophys.
Acta, 384, 430 (1975). The reaction medium con-tained 50 mM Tris-HC1, pH 7.5, 5 mM magnesium ace-tate, 250 ~.rg/ml 5'-Nucleotidase, 1 mM EGTA, and 0.15 ,uM 8- [H3] -cGMP. Unless otherwise indicated, the enzyme used was a human recombinant PDE5 (ICOS
Corp., Bothell, Washington).
Compounds of the invention were dissolved in DMSO finally present at 2o in the assay. The incubation time was 30 minutes during which the total substrate conversion did not exceed 30%.
The ICso values for the compounds examined were determined from concentration-response curves typically using concentrations ranging from 10 nM to 10 uM. Tests against other PDE enzymes using standard methodology showed that compounds of the invention are selective for the cGMP-specific PDE
enzyme.

Biological Data The compounds according to the present invention were typically found to exhibit an ICso value of less than 500 nM (i.e., 0.5 ,uM). In vitro test data for representative compounds of the inven-tion is given in the following table:
Table 1: In vitro Results (Examples) Example PDE5 ICSO y.lM) 1 0.044 3 0.241 4 1~ 0.2 5 1~ 0.49 6 0:23 7 l 1 8 .502 9 .261 11 .629 12 .03 13 .42 vs. bovine aorta The intermediates also exhibited a low ICso value of less than 900 nM, typically less than 1 uM
(i.e., 1000 nM), and often less than 0.5 ~M, as illustrated in the following summary of in vitro test data.

Table 2: In v itro Results (Intermediates) Intermediate PDE5 ICso (,t,~I) la 1' 0.85 1b 1~ 0 . 65 2 1~ 0.2 3 l~ 0.5 4 1' 0.8 5 1' 0.9 6 0.51 7a 1' 0.2 7b 1~ 0 . 7 8 0.76 9a 1' 0.47 9b 0.36 10 0.86 11 0.89 12 0.96 13a 0.27 13b 0.3 14 0.48 15a 0.29 15b 0.92 16a 0.53 16b 0.31 17a 0.38 17b 0.12 18 0.42 19 0.36 Table 2: In v itro Results (Intermediates) Intermediate PDES ICso (,i,~I) 20a 0.65 20b 0.34 21 0.68 22 0.68 23 0.8 24a 0.94 24b 0.92 25 0.59 26a 0.82 26b 0.54 27 0.83 28 0.22 29a 0.34 29b 0.23 30a 0.78 30b 0.42 31 0.43 32 0.33 33 0.86 34 0.23 35 0.16 36 0.09 37 0.62 38 0.77 39 0.65 vs. bovine aorta Obviously, many modifications and varia-tions of the invention as hereinbefore set forth can be made without departing from the spirit and scope thereof, and, therefore, only such limitations should be imposed as are indicated by the appended claims.

Claims (52)

WHAT IS CLAIMED IS:
1. A compound having a formula wherein R0, independently, is selected from the group consisting of halo, C1-6alkyl, aryl, heter-oaryl, C3-8cycloalkyl, C3-8heterocycloalkyl, C3-8cyclo-alkylQ, C(=O)R a, OC(=O)R a, C(=O)OR a, C1-4alkyleneNR a R b, C1-4alkyleneHet, C1-4alkyleneC(=O)OR a, C(=O)NR a SO2R c, C(=O)C1-4alkyleneHet, C(=O)NR a R b, C(=O)NR a R c, C(=O)-NR a C1-4alkyleneOR b, C(=O)NR a C1-4alkyleneHet, OR a, OC1-4-alkyleneC(=O)OR a, OC1-4alkyleneNR a R b, OC1-4alkyleneHet, OC1-4alkyleneOR a, OC1-4alkyleneNR a C(=O)OR b, NR a R b, NR a C1-4alkyleneNR a R b, NR aC(=O)R b, NR a C(=O)NR a R b, N(SO2C1-4alkyl)2, NR a(SO2C1-4alkyl), nitro, trifluoro-methyl, trifluoromethoxy, cyano, SO2NR a R b, SO2R a, SOR a, SR a, and OSO2CF3;
R1 is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, an optionally substituted C3-8cycloalkyl ring, an optionally substituted C3-8heterocycloalkyl ring, an optionally substituted bicyclic ring wherein the fused ring A is a 5- or 6-membered ring, saturated or partially or fully unsaturated, and comprises carbon atoms and optionally one to three heteroatoms selected from oxygen, sulfur, and nitrogen, hydrogen, C1-6alkyl, arylC1-3alkyl, C1-3-alkylenearyl, haloC1-6alkyl, C1-4alkyleneC(=O)OR a, C1-4alkyleneC(=O)NR a R b, C3-8cycloalkyl, C3-8cycloalkenyl, C3-8heterocycloalkenyl, C1-4alkyleneHet, C1-4alkylene-QR a, C3-6alkenyleneQR a, C1-4alkyleneQC1-4alkyleneQR a, and a spiro substituent having a structure R2 is selected from the group consisting of hydrogen, C1-6alkyl, C3-8cycloalkyl, C3-8heterocyclo-alkyl, C2-6alkenyl, C1-3alkylenearyl, arylC1-3alkyl, C(=O)R a, aryl, heteroaryl, C(=O)R a, C(=O)NR a R b, C(=O)-NR a R c, C(=S)NR a R b, C(=S)NR a R c, SO2R a, SO2NR a R b, S(=O)R a, S(=O)NR a R b, C(=O)NR a C1-4alkyleneOR a, C(=O)NR a C1-4alkyl-eneHet, C(=O)C1-4alkylenearyl, C(=O)C1-4alkylene-heteroaryl, C1-4alkylenearyl substituted with one or more of SO2NR a R b, NR aR b, C(=O)OR a, NR a SO2CF3, CN, NO2, C(=O)R a, OR a, C1-4alkyleneNR a R b, and OC1-4alkyleneNR a R b, C1-4alkyleneheteroaryl, C1-4alkyleneHet, C1-4alkyleneC-(=O)C1-4alkylenearyl, C1-4alkyleneC(=O)C1-4alkylene-heteroaryl, C1-4alkyleneC(=O)Het, C1-4alkyleneC(=O)-NR a R b, C1-4alkyleneOR a, C1-4alkyleneNR a C(=O)R a, C1-4alkyl-eneOC1-4alkyleneOR a, C1-4alkyleneNR a R b, C1-4alkyleneC-(=O)OR a, and C1-4alkyleneOC1-4alkyleneC(=O)OR a;

R3 is selected from the group consisting of C(=O)R b, C(=O)OR b, C(=O)NR a R b, C(=O)NR a R c, C(=S)NR a R b, C(=S)NR a R c, C(=O)Het, C(=O)NR a C1-4alkyleneOR a, C(=O)NR a C1-4alkyleneHet, C(=O)C1-4alkylenearyl, C(=O)C1-4alkyleneheteroaryl, C(=O)NR a C1-4alkylenearyl, C(=O)NR a C1-4alkyleneC3-8cycloalkyl, C(=O)NR b SO2R c, C(=O)NR a C1-4alkyleneOC1-6alkyl, C(=O)NR a C1-4alkylene-heteroaryl, NR a R c, NR a C(=O)R b, NR a C(=O)NR a R c, NR a(SO2C1-4alkyl), N(SO2C1-4alkyl)2, OR a, NR a C(=O)C1-4alkyleneN(R b)2, NR a C(=O)C1-4alkyleneC(=O)OR a, NR a(C=O)C1-3alkylenearyl, NR a C(=O)C1-3alkylene-C3-8heterocycloalkyl, NR a C(=O)C1-3alkyleneHet, and C(=O)NR a SO2R b;
R4 is selected from the group consisting of hydrogen, C1-6alkyl, aryl, heteroaryl, arylC1-3alkyl, C1-3alkylenearyl, C1-3alkyleneHet, C3-8cycloalkyl, and C3-8heterocycloalkyl;
X is selected from the group consisting of C(=O),(CH2)t C(=O), C(=O)C.ident.C, C(=O)C(R a)=C(R a), C(=S), SO, SO2, SO2C(R a)=CR a, CR a R b, CR a=CR a, C(=O)NR a, and C(=N-OR a);
Y is selected from the group consisting of R a, R d,(CH2)n C(=)R c, N(R b)(CH2)n R c, O(CH2)n R c, N(R b)C(=O)R c, C(=O)N(R a)(R c), N(R a)C(=0)R c, and N(R a)SO2R c;

R a is selected from the group consisting of hydrogen, C1-6alkyl, C3-6cycloalkyl, aryl, arylC1-3alkyl, C1-3alkylenearyl, heteroaryl, heteroarylC1-3alkyl, and C1-3alkyleneheteroaryl;
R b is selected from the group consisting of hydrogen, C1-6alkyl, C3-8cycloalkyl, C1-3alkyleneN(R d)2, aryl, arylC1-3alkyl, C1-3alkylenearyl, and heteroaryl;
R c is selected from the group consisting of hydrogen, C1-6alkyl, aryl, heteroaryl, arylC1-3alkyl, heteroarylC1-3alkyl, C1-3alkyleneN(R a)2, C1-6alkylene-aryl, C1-6alkyleneHet, haloC1-6alkyl, C3-8cycloalkyl, C3-8heterocycloalkyl, Het, C1-3alkyleneheteroaryl, C1-6alkyleneC(=O)OR a, and C1-3alkyleneC3-8heterocyclo-alkyl;
or R a and R c are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;
R4 is a 5- or 6-membered ring or a bicyclic fused ring system, saturated or partially or fully unsaturated, comprising carbon atoms and optionally one to three heteroatoms selected from oxygen, sulfur, and nitrogen, and optionally substituted with one or more R e or R f;
R e is selected from the group consisting of nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, Het, C1-6alkyl, C1-6alkyleneOR a, C(=O)R a, OC(=O)R a, C(=O)OR a, C1-4alkyleneHet, C1-4alkyleneC(=O)OR a, OC1-4alkyleneC(=O)OR a, C1-4alkyleneOC1-4alkyleneC(=O)OR a, C(=O)NR a SO2R f, C(=O)C1-4alkyleneHet, C1-4alkyleneNR a R b, C2-6alkenyleneNR a R b, C(=O)NR a R b, C(=O)NR a R g, C(=O)NR aC1-4alkyleneOR b, C(=O)NR a C1-4alkyleneHet, OR a, OC2-4alkyleneNR a R b, OC1-4alkyleneCH(OR a)CH2-NR a R b, OC1-4alkyleneHet, OC2-4alkyleneOR a, OC2-4alkyleneNR a C(=O)OR b, NR a R b, NR a C1-4alkyleneNR a R b, NR a C(=O)R b, NR a C(=O NR a R b, N(SO2C1-4alkyl)2, NR a(SO2C1-4alkyl), SO2NR a R b, and OSO2trifluoromethyl;
R f is selected from the group consisting of hydrogen, halogen, OR a, C1-6alkyl, nitro, and NR a R b;
or R e and R f are taken together to form a 3- or 4-membered alkylene or alkenylene chain component of a 5- or 6-membered ring, optionally containing at least one heteroatom;
R g is phenyl or C4-6cycloalkyl, optionally substituted with one or more halogen, C(=O)OR a, or OR a;
Q is O, S, or NR h;
B is O, S, or NR h;
C is O, S, or NR a;
D is CR a or N;
E is CR a, C(R a)2, or NR h; and R h is null or is selected from the group consisting of hydrogen, C1-6alkyl, aryl, heteroaryl, arylC1-3alkyl, heteroarylC1-3alkyl, C1-3alkylenearyl, and C1-3alkyleneheteroaryl;
Het represents a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1-4alkyl or C(=O)OR a;
n is 0 or 1;
q is 0, 1, 2, 3, or 4;
t is 1, 2, 3, or 4;
and pharmaceutically acceptable salts and solvates thereof.
2. The compound of claim 1 represented by the formula and pharmaceutically acceptable salts and hydrates thereof.
3. The compound of claim 1 wherein q is 0, or R0 is selected from the group consisting of aryl, Het, OR a, C(=O)OR a, C1-4alkyleneNR a R b, OC(=O)R a, C(=O)R a, NR a R b, C3-8cycloalkyl, C3-8cycloalkylQ, C(=O)NR a R b, and C(=O)NR a R c.
4. The compound of claim 1 wherein R1 is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, C1-4alkyleneQR a, C1-4alkyleneQC1-4alkyleneQR a, C3-8cyclo-alkyl, C3-8cycloalkenyl, C1-6alkyl,
5. The compound of claim 1 wherein R1 is the optionally substituted bicyclic ring system
6. The compound of claim 5 wherein R1 is and wherein p is an integer 1 or 2, and G, independ-ently, are C(R a)2, O, S, or NR a.
7. The compound of claim 1 wherein R1 is selected from the group consisting of -CH2OR a, -CH2OCH2OR a,
8. The compound of claim 1 wherein R2 is selected from the group consisting of hydrogen, aryl, heteroaryl, OR a, NR a R b, NR a R c, C1-4alkyleneHet, C1-4alkyleneheteroaryl, C1-4alkylenearyl, C1-4alkyleneC-(=O)C1-4alkylenearyl, C1-4alkyleneC(=O)OR a, C1-4alkyl-eneC(=O)NR a R b, C1-4alkyleneC(=O)NR a R c, C1-4alkyleneC-(=O)Het, C1-4alkyleneNR a R b, C1-4alkyleneNR a R c, C1-4alkyl-eneNR a C(=O)R a, and C1-4alkyleneOC1-4alkyleneOR a.
9. The compound of claim 8 wherein R2 is selected from the group consisting of hydrogen; C1-4-alkyleneheteroaryl, wherein the heteroaryl group is selected from the group consisting of benzimidazole, a triazole, and imidazole; C1-4alkyleneHet, wherein Het is selected from the group consisting of piper-azine, morpholine, pyrrolidine, pyrrolidone, tetra-hydrofuran, piperidine, C1-4alkyleneC6H5, optionally substituted with one to three groups selected from the group consisting of C(=O)OR a, NR a R b, NR a SO2CF3, SO2NR a R b, CN, OR a, C(=O)R a, C1-4alkyleneNR a R b, nitro, OC1-4alkylenearyl, and OC1-4-alkyleneNR a R b; C1-4alkyleneC(=O)benzyl; C1-4alkylene-C(=O)OR a; C1-4alkyleneC(=O)NR a R b; C1-4alkyleneC(=O)NR a R c;
C1-4alkyleneHet; NR a R b; OH; OC1-4alkyl; C6H5; C1-4alkyl-eneNR a R b; C1-4alkyleneOR a; C1-4alkyleneNHC(=O)R a; and C1-4alkyleneOC1-4alkyleneOR a.
10. The compound of claim 8 wherein R2 is selected from the group consisting of C1-6alkyl, C(=O)OR a, C(=O)R a, hydrogen, C(=O)NR a C1-4alkyleneHet, C(=O)NR a R c, aryl, and heteroaryl.
11. The compound of claim 1 wherein R3 is selected from the group consisting of C(=O)OR a, C(=O)R a, C(=O)NR a C1-4alkyleneOC1-6alkyl, C(=O)NR a C1-4-alkyleneC3-8cycloalkyl, C(=O)Het, C(=O)NR a C1-4alkyleneheteroaryl, C(=O)NR a C1-4alkylene-aryl, C(=O)NR a C1-4alkyleneHet, C(=O)NR a R c, and C(=S)NR a R c.
12. The compound of claim 1 wherein R4 is selected from the group consisting of hydrogen, C1-6alkyl, aryl, and heteroaryl.
13. The compound of claim 1 wherein n is 0, or X is selected from the group consisting of C(=O), C(=O)CR a=CR a, (CH2)t C=O, C(=S) and C(=N-OR a).
14. The compound of claim 1 wherein Y is selected from the group consisting of R a, R d, N(R b)(CH2)n R c, N(R b)C(=O)R c, and N(R a)C(=O)R c.
15. The compound of claim 1 wherein q is 0, or R0 is selected from the group consisting of halo, methyl, trifluoromethyl, and trifluoromethyl;
R1 is selected from the group consisting of R2 is selected from the group consisting of hydrogen, C1-6alkyl, C (=O) NRaRc, and C1-4alkyleneHet; R3 is selected from the group consisting of C (=O) OC2H5, C (=O) OCH3, C (=O) NHCH2C6H5, C (=O) NH (CH2) 2C6H5, C (=O) NHC6H5, C (=O) NH2, C (=O) N (CH3) 2, C (=S) N (CH3) 2, C (=O) NH (CH2) 2CH3, C (=O) N (CH2) 3CH3, C (=O) NHCH3, C (=O) NHCH (CH3) 2, C (=O) NH (CH2) 3OCH3, and R4 is hydrogen or C1-6alkyl.
16. The compound of claim 1 wherein q is 0, R2 is hydrogen, and R4 is hydrogen or methyl.
17. A compound selected from the group consisting of and pharmaceutically acceptable salts and solvates thereof.
18. A pharmaceutical composition com-prising a compound of claim 1, together with a pharmaceutically acceptable diluent or carrier.
19. A method of treating a male or female animal in the treatment of a condition where inhi-bition of a cGMP-specific PDE is of a therapeutic benefit comprising treating said animal with an effective amount of a pharmaceutical composition comprising a compound of claim 1, together with a pharmaceutically acceptable diluent or carrier.
20. The method of claim 19 wherein the condition is male erectile dysfunction.
21. The method of claim 20 wherein the treatment is an oral treatment.
22. The method of claim 19 wherein the condition is female arousal disorder.
23. The method of claim 22 wherein the treatment is an oral treatment.
24. The method of claim 19 wherein the condition is selected from the group consisting of stable angina, unstable angina, variant angina, hypertension, pulmonary hypertension, chronic ob-structive pulmonary disease, malignant hypertension, pheochromocytoma, acute respiratory distress syn-drome, congestive heart failure, acute renal fail-ure, chronic renal failure, atherosclerosis, a con-dition of reduced blood vessel patency, a peripheral vascular disease, a vascular disorder, thrombocy-themia, an inflammatory disease, myocardial infarc-tion, stroke, bronchitis, chronic asthma, allergic asthma, allergic rhinitis, glaucoma, peptic ulcer, a gut motility disorder, postpercutaneous transluminal coronary angioplasty, carotid angioplasty, post-bypass surgery graft stenosis, osteoporosis, preterm labor, benign prostatic hypertrophy, and irritable bowel syndrome.
25. A method of treating a condition where inhibition of a cGMP-specific PDE is of thera-peutic benefit, in a human or a nonhuman animal body, comprising administering to said body a thera-peutically effective amount of a compound of claim 1.
26. A method for the curative or prophyl-actic treatment of male erectile dysfunction or female arousal disorder, comprising administration of an effective dose of a compound of claim 1, and pharmaceutically acceptable salts and solvates thereof, to an animal.
27. Use of a compound of claim 1 for the manufacture of a medicament for the curative or prophylactic treatment of a condition where inhibi-tion of a cGMP-specific PDE is of a therapeutic benefit.
28. A compound having a formula wherein R0, independently, is selected from the group consisting of halo, C1-6alkyl, aryl, heter-oaryl , C3-8cycloalkyl , C3-8heterocycloalkyl , C3-8cyclo-alkylQ, C(=O)R a, OC(=O)R a, C(=O)OR a, C1-4alkyleneNR a R b, C1-4alkyleneHet, C1-4alkyleneC(=O)OR a, C(=O)NR a SO2R c, C(=O) C1-4alkyleneHet, C(=O) NR a R b, C(=O)NR a R c, C(=O)-NR a C1-4alkyleneOR b, C(=O)NR a C1-4alkyleneHet, OR a, OC1-4-alkyleneC(=O)OR a, OC1-4alkyleneNR a R b, OC1-4alkyleneHet, OC1-4alkyleneOR a, OC1-4alkyleneNR a C(=O)OR b, NR a R b, NR a C1-4alkyleneNR a R b, NR a C (=O)R b, NR a C (=O)NR a R b, N(SO2C1-4alkyl)2, NR a(SO2C1-4alkyl), nitro, trifluoro-methyl, trifluoromethoxy, cyano, SO2NR a R b, SO2R a, SOR a, SR a, and OSO2CF3;

R1 is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, an optionally substituted C3-8cycloalkyl ring, an optionally substituted C3-8heterocycloalkyl ring, an optionally substituted bicyclic ring wherein the fused ring A is a 5- or 6-membered ring, saturated or partially or fully unsaturated, and comprises carbon atoms and optionally one to three heteroatoms selected from oxygen, sulfur, and nitrogen, hydrogen, C1-6alkyl, arylC1-3alkyl, C1-3-alkylenearyl, haloC1-6alkyl, C1-4alkyleneC(=O)OR a, C1-4alkyleneC(=O)NR a R b, C3-8cycloalkyl, C3-8cycloalkenyl, C3-8heterocycloalkenyl, C1-4alkyleneHet, C1-4alkylene-QR a, C2-6alkenyleneQR a, C1-4alkyleneQC1-4alkyleneQR a, and a spiro substituent having a structure R2 is selected from the group consisting of hydrogen, C1-6alkyl , C3-8cycloalkyl , C3-8heterocyclo-alkyl, C2-6alkenyl, C1-3alkylenearyl, arylC1-3alkyl, C(=O)R a, aryl, heteroaryl, C(=O)R a, C(=O)NR a R b, C(=O)-NR a R c , C(=S)NR a R b, C(=S)NR a R c , SO2R a , SO2NR a R b , S(=O)R a, S(=O)NR a R b, C(=O)NR a C1-4alkyleneOR a, C(=O)NR a C1-4alkyl-eneHet, C(=O)C1-4alkylenearyl, C(=O)C1-4alkylene-heteroaryl, C1-4alkylenearyl substituted with one or more Of SO2NR a R b, NR a R b, C(=O)OR a, NR a SO2CF3, CN, NO2, C(=O)R a, OR a, C1-4alkyleneNR a R b, and OC1-4alkyleneNR a R b, C1-4alkyleneheteroaryl, C1-4alkyleneHet, C1-4alkyleneC-(=O)C1-4alkylenearyl, C1-4alkyleneC (=O) C1-4alkylene-heteroaryl, C1-4alkyleneC(=O) Het, C1-4alkyleneC(=O)-NR a R b, C1-4alkyleneOR a, C1-4alkyleneNR a C(=O)R a, C1-4alkyl-eneOC1-4alkyleneOR a, C1-4alkyleneNR a R b, C1-4alkyleneC-(=O)OR a, and C1-4alkyleneOC1-4alkyleneC(=O)OR a;
R3 is selected from the group consisting of C(=O)R b, C(=O)OR b, C(=0)NR a R b, C(=O)NR a R c, C(=S) NR a R b, C(=S)NR a R c, C(=O)Het, C(=O)NR a C1-4alkyleneOR a, C(=O)-NR a C1-4alkyleneHet, C(=O)C1-4alkylenearyl, C(=O)C1-4-alkyleneheteroaryl, C(=O)NR a C1-4alkylenearyl, C(=O)-NR a C1-4alkyleneC3-8cycloalkyl, C(=O)NR b SO2R c, C(=O)-NR a C1-4alkyleneOC1-6alkyl, C(=O)NR a C1-4alkylenehetero-aryl, NR a R c, NR a C(=O)R b, NR a C(=O)NR a R c, NR a(SO2C1-4-alkyl), N(SO2C1-4alkyl)2; OR a, NR a C(=O)C1-4alkylene-N(R b)2, NR a C(=O)C1-4alkyleneC(=O)OR a, NR a(C=O)C1-3-alkylenearyl, NR a C(=O)C1-3alkyleneC3-8heterocycloalkyl, NR a C(=O)C1-3alkyleneHet, and C(=O)NR a SO2R b;
R4 is selected from the group consisting of hydrogen, C1-6alkyl, aryl , heteroaryl , arylC1-3alkyl, C1-3alkylenearyl, C1-3alkyleneHet, C3-8cycloalkyl, and C3-8heterocycloalkyl;
R a is selected from the group consisting of hydrogen, C1-6alkyl, C3-6cycloalkyl, aryl, arylC1-3alkyl, C1-3alkylenearyl, heteroaryl, heteroarylC1-3alkyl, and C1-3alkyleneheteroaryl;
R b is selected from the group consisting of hydrogen, C1-6alkyl,. C3-8cycloalkyl, C1-3alkyleneN(R a)2.
aryl , arylC1-3alkyl, C1-3alkylenearyl, and heteroaryl;
R c is selected from the group consisting of hydrogen, C1-6alkyl, aryl, heteroaryl, arylC1-3alkyl, heteroarylC1-3alkyl, C1-3alkyleneN(R a)2, C1-6alkylene-aryl, C1-6alkyleneHet, haloC1-6alkyl, C3-8cycloalkyl, C3-8heterocycloalkyl, Het, C1-3alkyleneheteroaryl, C1-6alkyleneC(=O)OR a, and C1-3alkyleneC3-8heterocyclo-alkyl;
or R a and R c are taken together to form a 5- or 6-membered ring, optionally containing at least one heteroatom;
Q is O, S, or NR h;
B is O, S, or NR h;
C is O, S, or NR a;
D is CR a or N;
E is CR a, C(R a)2, or NR h; and R h is null or is selected from the group consisting of hydrogen, C1-6alkyl, aryl, heteroaryl, arylC1-3alkyl, heteroarylC1-3alkyl, C1-3alkylenearyl, and C1-3alkyleneheteroaryl;
Het represents a 5- or 6-membered heterocyclic ring, saturated or partially or fully unsaturated, containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and optionally substituted with C1-4alkyl or C(=O)OR a;
n is 0 or 1;
q is 0, 1, 2, 3, or 4;
t is 1, 2, 3, or 4;

and pharmaceutically acceptable salts and solvates thereof.
29. The compound of claim 28 represented by the formula and pharmaceutically acceptable salts and hydrates thereof.
30. The compound of claim 28 wherein q is 0, or R0 is selected from the group consisting of aryl, Het, OR a, C(=O)OR a, C1-4alkyleneNR a R b, OC(=O)R a, C(=O)R a, NR a R b, C3-8cycloalkyl, C3-8cycloalkylQ, C(=O)NR a R b, and C(=O)NR a R c.
31. The compound of claim 28 wherein R1 is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, C1-4alkyleneQR a, C1-4alkyleneQC1-4alkyleneQR a, C3-8cycloalkyl , C3-8cycloalkenyl , C1-6alkyl,
32. The compound of claim 28 wherein R1 is the optionally substituted bicyclic ring system
33. The compound of claim 32 wherein R1 is and wherein p is an integer 1 or 2, and G, indepen-dently, are C(R a)2, O, S, or NR a.
34. The compound of claim 28 wherein R1 is selected from the group consisting of -CH2OR a, -CH2OCH2OR a
35. The compound of claim 28 wherein R2 is selected from the group consisting of aryl, hydro-gen, heteroaryl, OR a, NR a R b, NR a R c, C1-4alkyleneHet, C1-4alkyleneheteroaryl, C1-4alkylenearyl, C1-4alkyleneC-(=O)C1-4alkylenearyl, C1-4alkyleneC(=O)OR a, C1-4alkyl-eneC(=O)NR a R b, C1-4alkyleneC(=O)NR a R c, C1-4alkyleneC-(=O)Het, C1-4alkyleneNR a R b, C1-4alkyleneNR a R c, C1-4alkyl-eneNR a C(=O)R a, and C1-4alkyleneOC1-4alkyleneOR a.
36. The compound of claim 28 wherein R2 is selected from the group consisting of hydrogen; C1-4-alkyleneheteroaryl, wherein the heteroaryl group is selected from the group consisting of benzimidazole, a triazole, and imidazole; C1-4alkyleneHet, wherein Het is selected from the group consisting of piperazine, morpholine, pyrrolidine, pyrrolidone, tetrahydrofuran, piperidine, C1-4alkyleneC6H5, optionally substituted with one to three groups selected from the group consisting of C(=O)OR a, NR a R b, NR a SO2CF3, SO2NR a R b, CN, OR a, C(=O)R a, C1-4alkyleneNR a R b, nitro, OC1-4alkylenearyl, and OC1-4alkyleneNR a R b; C1-4alkyleneC(=O)benzyl; C1-4alkyl-eneC(=O)OR a; C1-4alkyleneC(=O)NR a R b; C1-4alkyleneC(=O)-NR a R c; C1-4alkyleneHet; NR a R b; OH; OC1-4alkyl; C6H5;
C1-4alkyleneNR a R b; C1-4alkyleneOR a; C1-4alkyleneNHC-(=O)R a; and C1-4alkyleneOC1-4alkyleneOR a.
37. The compound of claim 28 wherein R2 is selected from the group consisting of C1-6alkyl, C(=O)OR a, C(=O)R a, hydrogen, C(=O)NR a C1-4alkyleneHet, C(=O)NR a R c, aryl, and heteroaryl.
38. The compound of claim 28 wherein R3 is selected from the group consisting of C(=O)OR a, C(=O)R a, C(=O)NR a C1-4alkyleneOC1-6alkyl, C(=O)NR a C1-_4-alkyleneC3-8cycloalkyl, C(=O)Het, C(=O)NR a C1-4alkyleneheteroaryl, C(=O)NR a C1-4alkylene-aryl, C(=O)NR a C1-4alkyleneHet, C(=O)NR a R c, and C(=S)NR a R c.
39. The compound of claim 28 wherein R4 selected from the group consisting of hydrogen, C1-6alkyl, aryl, and heteroaryl.
40. The compound of claim 1 wherein q is 0, or R0 is selected from the group consisting of halo, methyl, trifluoromethyl, and trifluoromethyl;
R1 is selected from the group consisting of , and R2 is selected from the group consisting of hydrogen, C1-6alkyl, C(=O)NR a R c, and C1-4alyleneHet; R3 is selected from the group consisting of C(=O)OC2H5, C(=O)OCH3, C(=O)NHCH2C6H5, C(=O)NH(CH2)2C6H5, C(=O)NHC6H5, C(=O)NH2, C(=O)N(CH3)2, C(=S)N(CH3)2, C(=O)NH(CH2)2CH3, C(=O)N(CH2)3CH3, C(=O)NHCH3, C(=O)NHCH(CH3)2, C(=O)NH(CH2)3OCH3, C (=O)NH (CH2) , C (=O)NHCH2 , C (=O)NH (CH2)2 , C (=O)NHCH2 C (=O)NH (CH2)2 and R4 is hydrogen and C1-6alkyl.
41. The compound of claim 28 wherein q is 0, R2 is hydrogen, and R4 is hydrogen or methyl.
42. A compound selected from the group consisting of and pharmaceutically acceptable salts and solvates thereof.
43. A pharmaceutical composition com-prising a compound of claim 28, together with a pharmaceutically acceptable diluent or carrier.
44. A method of treating a male or female animal in the treatment of a condition where inhi-bition of a cGMP-specific PDE is of a therapeutic benefit comprising treating said animal with an effective amount of a pharmaceutical composition comprising a compound of claim 28, together with a pharmaceutically acceptable diluent or carrier.
45. The method of claim 44 wherein the condition is male erectile dysfunction.
46. The method of claim 44 wherein the condition is female arousal disorder.
47. The method of claim 45 wherein the treatment is an oral treatment.
48. The method of claim 46 wherein the treatment is an oral treatment.
49. The method of claim 44 wherein the condition is selected from the group consisting of stable angina, unstable angina, variant angina, hypertension, pulmonary hypertension, chronic ob-structive pulmonary disease, malignant hypertension, pheochromocytoma, acute respiratory distress syn-drome, congestive heart failure, acute renal fail-ure, chronic renal failure, atherosclerosis, a con-dition of reduced blood vessel patency, a peripheral vascular disease, a vascular disorder, thrombocy-themia, an inflammatory disease, myocardial infarc-tion, stroke, bronchitis, chronic asthma, allergic asthma, allergic rhinitis, glaucoma, peptic ulcer, a gut motility disorder, postpercutaneous transluminal coronary angioplasty, carotid angioplasty, post-bypass surgery graft stenosis, osteoporosis, preterm labor, benign prostatic hypertrophy, and irritable bowel syndrome.
50. A method of treating a condition where inhibition of a cGMP-specific PDE is of thera-peutic benefit, in a human or a nonhuman animal body, comprising administering to said body a thera-peutically effective amount of a compound of claim 28.
51. A method for the curative or prophyl-actic treatment of male erectile dysfunction or female arousal disorder, comprising administration of an effective dose of a compound of claim 28, and pharmaceutically acceptable salts and solvates thereof, to an animal.
52. Use of a compound of claim 28 for the manufacture of a medicament for the curative or prophylactic treatment of a condition where inhibi-tion of a cGMP-specific PDE is of a therapeutic benefit.
CA2441792A 2001-04-25 2002-04-02 Carboline derivatives as inhibitors of phosphodiesterase 5 (pdes) for the treatment of cardiovascular diseases and erectile dysfunction Expired - Fee Related CA2441792C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28673001P 2001-04-25 2001-04-25
US60/286,730 2001-04-25
PCT/US2002/010367 WO2002088123A1 (en) 2001-04-25 2002-04-02 Chemical compounds

Publications (2)

Publication Number Publication Date
CA2441792A1 CA2441792A1 (en) 2002-11-07
CA2441792C true CA2441792C (en) 2010-08-03

Family

ID=23099912

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2441792A Expired - Fee Related CA2441792C (en) 2001-04-25 2002-04-02 Carboline derivatives as inhibitors of phosphodiesterase 5 (pdes) for the treatment of cardiovascular diseases and erectile dysfunction

Country Status (9)

Country Link
US (1) US7115621B2 (en)
EP (1) EP1383765B1 (en)
JP (1) JP4216606B2 (en)
AT (1) ATE348101T1 (en)
CA (1) CA2441792C (en)
DE (1) DE60216745T2 (en)
ES (1) ES2278956T3 (en)
MX (1) MXPA03009750A (en)
WO (1) WO2002088123A1 (en)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI329111B (en) 2002-05-24 2010-08-21 X Ceptor Therapeutics Inc Azepinoindole and pyridoindole derivatives as pharmaceutical agents
JP4671123B2 (en) 2003-06-23 2011-04-13 小野薬品工業株式会社 New tricyclic heterocyclic compounds
EA016575B1 (en) * 2004-03-15 2012-06-29 ПиТиСи ТЕРАПЬЮТИКС, ИНК. Carboline derivatives useful in the inhibition of angiogenesis
US8076352B2 (en) 2004-03-15 2011-12-13 Ptc Therapeutics, Inc. Administration of carboline derivatives useful in the treatment of cancer and other diseases
US7767689B2 (en) * 2004-03-15 2010-08-03 Ptc Therapeutics, Inc. Carboline derivatives useful in the treatment of cancer
US8076353B2 (en) 2004-03-15 2011-12-13 Ptc Therapeutics, Inc. Inhibition of VEGF translation
US8063214B2 (en) * 2004-10-28 2011-11-22 Dr. Reddy's Laboratories Limited Polymorphic forms of tadalafil
AR051780A1 (en) * 2004-11-29 2007-02-07 Japan Tobacco Inc FUSIONED RING COMPOUNDS CONTAINING NITROGEN AND USING THEMSELVES
ES2459742T3 (en) * 2004-12-22 2014-05-12 Ono Pharmaceutical Co., Ltd. Tricyclic compound and use thereof
EP1747779A1 (en) 2005-07-28 2007-01-31 Laboratorios Del Dr. Esteve, S.A. Tetrahydro-b-carbolin-sulfonamide derivatives as 5-HT6 ligands
KR20080047375A (en) * 2005-07-29 2008-05-28 콘서트 파마슈티컬즈, 인크. Novel pharmaceutical compounds
WO2007020521A1 (en) * 2005-08-16 2007-02-22 Pharmacia & Upjohn Company Llc Pyridoyrazinones as pde-5 inhibitors
WO2007110734A1 (en) * 2006-03-24 2007-10-04 Glenmark Pharmaceuticals Limited Process for the preparation of tadalafil
WO2009038764A1 (en) * 2007-09-20 2009-03-26 D2E, Llc Fluoro-containing derivatives of hydrogented pyrido[4,3-b]indoles with neuroprotective and cognition enhancing properties, process for preparing, and use
JP5501251B2 (en) 2008-01-11 2014-05-21 アルバニー モレキュラー リサーチ, インコーポレイテッド (1-Azinone) substituted pyridoindoles as MCH antagonists
US8618299B2 (en) 2009-07-01 2013-12-31 Albany Molecular Research, Inc. Azinone-substituted azapolycycle MCH-1 antagonists, methods of making, and use thereof
US9073925B2 (en) * 2009-07-01 2015-07-07 Albany Molecular Research, Inc. Azinone-substituted azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
EP2448585B1 (en) * 2009-07-01 2014-01-01 Albany Molecular Research, Inc. Azinone-substituted azepino[b]indole and pyrido-pyrrolo-azepine mch-1 antagonists, methods of making, and use thereof
US8629158B2 (en) 2009-07-01 2014-01-14 Albany Molecular Research, Inc. Azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US8993765B2 (en) 2010-12-21 2015-03-31 Albany Molecular Research, Inc. Tetrahydro-azacarboline MCH-1 antagonists, methods of making, and uses thereof
WO2012088038A2 (en) 2010-12-21 2012-06-28 Albany Molecular Research, Inc. Piperazinone-substituted tetrahydro-carboline mch-1 antagonists, methods of making, and uses thereof
EP2687531B1 (en) 2011-03-18 2016-07-13 ONO Pharmaceutical Co., Ltd. Tetrahydrocarboline derivative
CN102391323B (en) * 2011-10-18 2013-09-25 首都医科大学 Tetrahydro-beta-carboline derivative, preparation method thereof and use thereof
TW201710258A (en) 2015-06-23 2017-03-16 碩騰服務公司 Carboline antiparasitics

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES273769A1 (en) * 1961-01-18 1962-06-01 Geigy Ag J R Process for the preparation of new -carboline-carboxylic acid amide derivatives
JPS5543475B2 (en) * 1972-09-14 1980-11-06
US4336260A (en) * 1977-12-08 1982-06-22 The Dow Chemical Company Method and compositions using 1-aryl-1,2,3,4-tetrahydro-β-carboline-3-carboxylic acid for treating depression
JPS5936676A (en) * 1982-08-20 1984-02-28 Tanabe Seiyaku Co Ltd Novel preparation of beta-carboline
FR2663935A1 (en) * 1990-06-27 1992-01-03 Adir NOVEL 1,2,3,4,5,6-HEXAHYDROAZEPINO [4,5-B] INDOLES AND 1,2,3,4-TETRAHYDROBETHACARBOLINES, PROCESSES FOR PREPARING THEM AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
GB9401090D0 (en) * 1994-01-21 1994-03-16 Glaxo Lab Sa Chemical compounds
GB9514473D0 (en) * 1995-07-14 1995-09-13 Glaxo Lab Sa Chemical compounds
AU710079B2 (en) * 1996-04-04 1999-09-16 F. Hoffmann-La Roche Ag Use of derivatives of tetrahydro-beta-carbolines as antimetastatic agents
AU711885B2 (en) * 1996-05-10 1999-10-21 Icos Corporation Carboline derivatives
WO1997043267A1 (en) 1996-05-11 1997-11-20 Kings College London Pyrazines
US6043252A (en) * 1997-05-05 2000-03-28 Icos Corporation Carboline derivatives
EP1114048A1 (en) * 1998-09-16 2001-07-11 Agnès Bombrun Carboline derivatives as cgmp phosphodiesterase inhibitors
DZ3179A1 (en) * 1999-08-03 2001-02-08 Lilly Icos Llc Pharmaceutical compositions based on beta-carboline.
UA74826C2 (en) * 2000-05-17 2006-02-15 Ortho Mcneil Pharm Inc ?-carboline derivatives as phosphodiesterase inhibitors
CA2411008C (en) * 2000-06-07 2006-04-11 Lilly Icos Llc Derivatives of 2,3,6,7,12,12a-hexahydropyrazino[1',2':1,6]pyrido[3,4-b]indole-1,4-dione
EP1289990A1 (en) 2000-06-08 2003-03-12 Lilly Icos LLC Tetracyclic diketopierazine compounds as pdev inhibitors
US6858620B2 (en) * 2000-10-03 2005-02-22 Lilly Icos, Llc Condensed pyridoindole derivatives
US6878711B2 (en) 2000-11-06 2005-04-12 Lilly Icos Llc Indole derivatives as PDE5-inhibitors

Also Published As

Publication number Publication date
US20040147542A1 (en) 2004-07-29
MXPA03009750A (en) 2004-06-30
JP2004532852A (en) 2004-10-28
WO2002088123A1 (en) 2002-11-07
EP1383765B1 (en) 2006-12-13
DE60216745D1 (en) 2007-01-25
CA2441792A1 (en) 2002-11-07
EP1383765A1 (en) 2004-01-28
ES2278956T3 (en) 2007-08-16
ATE348101T1 (en) 2007-01-15
JP4216606B2 (en) 2009-01-28
US7115621B2 (en) 2006-10-03
DE60216745T2 (en) 2007-05-16

Similar Documents

Publication Publication Date Title
CA2441792C (en) Carboline derivatives as inhibitors of phosphodiesterase 5 (pdes) for the treatment of cardiovascular diseases and erectile dysfunction
EP1360186B1 (en) Carboline derivatives
EP1335923B1 (en) Condensed pyridoindole derivatives
CA2411008C (en) Derivatives of 2,3,6,7,12,12a-hexahydropyrazino[1&#39;,2&#39;:1,6]pyrido[3,4-b]indole-1,4-dione
EP1313736A2 (en) Condensed pyrazindione derivatives
EP1335917B1 (en) Condensed pyridoindole derivatives
EP1294721B1 (en) Cyclic gmp-specific phosphodiesterase inhibitors
EP1392688B1 (en) Pyrazino[1&#39;,2&#39;:1,6]-pyrido[3,4-b] indole-1,4-dione derivatives
CA2445460A1 (en) Carboline derivatives as pde-5 inhibitors
CA2427573C (en) Indole derivatives as pde5-inhibitors
EP1366049B1 (en) Pirazino¬1&#39;2&#39;:1,6|pyrido¬3,4-b|indole derivatives
CA2445189C (en) Carboline derivatives as pdev inhibitors

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20170403